Next Article in Journal
Polyunsaturated Fatty Acid-Enriched Lipid Fingerprint of Glioblastoma Proliferative Regions Is Differentially Regulated According to Glioblastoma Molecular Subtype
Next Article in Special Issue
Molecular Mechanisms of Pregnancy-Related Vascular Remodeling and Pregnancy Complications
Previous Article in Journal
Use of Nanocellulose to Produce Water-Based Conductive Inks with Ag NPs for Printed Electronics
Previous Article in Special Issue
Defective Uteroplacental Vascular Remodeling in Preeclampsia: Key Molecular Factors Leading to Long Term Cardiovascular Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modeling Trophoblast Cell-Guided Uterine Spiral Artery Transformation in the Rat

by
Vinay Shukla
1,* and
Michael J. Soares
1,2,3,*
1
Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
2
Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
3
Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(6), 2947; https://doi.org/10.3390/ijms23062947
Submission received: 4 February 2022 / Revised: 3 March 2022 / Accepted: 4 March 2022 / Published: 9 March 2022

Abstract

:
The rat possesses hemochorial placentation with deep intrauterine trophoblast cell invasion and trophoblast-guided uterine spiral artery remodeling, which resembles human placentation. Uterine spiral arteries are extensively remodeled to deliver sufficient supply of maternal blood and nutrients to the developing fetus. Inadequacies in these key processes negatively impact fetal growth and development. Recent innovations in genome editing combined with effective phenotyping strategies have provided new insights into placental development. Application of these research approaches has highlighted both conserved and species-specific features of hemochorial placentation. The review provides foundational information on rat hemochorial placental development and function during physiological and pathological states, especially as related to the invasive trophoblast cell-guided transformation of uterine spiral arteries. Our goal is to showcase the utility of the rat as a model for in vivo mechanistic investigations targeting regulatory events within the uterine-placental interface.

1. Introduction

The placenta is an extraembryonic structure permitting survival of the fetus within the female reproductive tract [1,2]. Among mammals, placentas exhibit differences in their structural organization and vary in their connectivity to their maternal host [3]. Hemochorial placentation is the most invasive form of placentation [3,4]. Trophoblast cells, the specialized cell lineage of the placenta, have a range of functions that ensure the growth and maturation of the fetus [5]. Among these functions is the remodeling of uterine spiral arteries that supply maternal blood to the placenta [5,6,7]. Restructuring uterine spiral arteries at the uterine-placental interface is a multifaceted process involving several cell types. In addition to invasive trophoblast cells, maternal immune cells, including natural killer (NK) cells and macrophages, contribute to uterine spiral artery remodeling [5,8]. In humans, inadequate uterine spiral artery transformation in early pregnancy results in insufficient perfusion of the placenta and fetus and may lead to pregnancy-related disorders such as preeclampsia, fetal growth restriction (FGR), and preterm delivery [9,10]. Mouse, rat, rabbit, human, some non-human primates, and bats are among species possessing a hemochorial placenta [11]. The rat shares deep hemochorial placentation and trophoblast-directed uterine spiral artery restructuring with humans and is an experimentally tractable model [4,12,13].
In this review, we discuss the rat as an experimental model for investigations directed toward elucidation of mechanisms underlying the establishment of the uterine-placental interface. We present a framework for the experimental interrogation of the invasive trophoblast cell lineage and trophoblast-guided uterine spiral artery remodeling. Findings that highlight gene regulatory networks contributing to the formation of the uterine-placental interface of the rat as a model system for studying conserved physiologically relevant mechanisms controlling hemochorial placentation are emphasized in the review. Some data from other model systems are presented to support observations in the rat.

2. Organization of the Rat Placentation Site

We will start this section with comments regarding the relationship between maternal tissues and the placenta. The “placenta” is of extraembryonic origin, thus descriptors such as fetal or maternal placenta are not appropriate. Alternatively, the “placentation site” contains both maternal and extraembryonic contributions. Maternal and extraembryonic contributions to the placentation site are essential for a successful pregnancy. Most notably, the maternal uterine stroma transforms into the multi-functional decidua, uterine vasculature is re-engineered to facilitate the flow of nutrients to the conceptus, and maternal immune cells are recruited and/or redistributed within the uterus proximal to the developing placenta [14,15,16]. The placenta is defined by the behavior of its constituent trophoblast cells.
Based on established nomenclature, the rat placenta can be defined based on its histological structure: hemotrichorial, shape: discoid, and invasiveness: deep [11,12,13,17,18,19,20] (Figure 1). The human placenta is similar to the rat placenta in its shape and invasiveness, but its histological structure is distinct and characterized as hemomonochorial. The genesis of the placenta occurs at the initial differentiation event of the embryo when the trophoblast cell lineage is established [21]. Trophectoderm which constitutes the outer cell layer of the blastocyst is the source of trophoblast cell progenitors contributing to the formation of extraembryonic ectoderm and subsequently chorionic ectoderm and the ectoplacental cone [22]. These latter structures are precursors to the two major compartments of the rat placenta known as the labyrinth zone and the junctional zone [23,24]. In the literature, the junctional zone has also been referred to as the basal zone, trophospongiosum, spongiotrophoblast, or spongy region.
Epithelial (trophoblast)-mesenchymal cell interactions dictate placental morphogenesis and formation of the labyrinth zone. The labyrinth zone arises from the interaction of chorionic ectoderm with allantoic mesoderm and is the site of the three trophoblast cell layers comprising the hemotrichorial histological structure defining the rat placenta [24]. Trophoblast progenitor cell populations within the labyrinth zone fuse to form two syncytiotrophoblast layers or alternatively, undergo endoreduplication and differentiate into trophoblast giant cells [24,25,26]. Syncytiotrophoblast layers importantly contribute to the placental barrier, regulating maternal-fetal nutrient and waste exchange [27,28], whereas trophoblast giant cells possess an endocrine function, which notably includes the secretion of placental lactogens [25].
The ectoplacental cone gives rise to the junctional zone, a heterogeneous epithelial cell structure containing trophoblast cell-lined channels for blood delivery to the labyrinth zone [23,25,29]. The junctional zone develops between the labyrinth zone and the uterine stroma. Several differentiated cell types arise within the junctional zone and include trophoblast giant cells, spongiotrophoblast cells, glycogen trophoblast cells, and invasive trophoblast cells [5,13]. Progenitor cell-progression to differentiated cell lineages within the junctional zone has not been determined. Cellular constituents of the junctional zone contribute to the maternal endocrine milieu (trophoblast giant cells, spongiotrophoblast cells), placental energy reserve (glycogen trophoblast cells), and to the transformation of the uterus (invasive trophoblast cells) [25,30,31,32]. Invasive trophoblast cells penetrate the mesometrial side of the uterus where uterine spiral arteries/arterioles are situated and extend into a region previously termed the “metrial gland” [33] but now referred to as the “uterine-placental interface” [32]. Invasive trophoblast cells can be categorized by their route of entry into the uterus. Endovascular invasive trophoblast cells enter the vasculature and displace endothelial cells, whereas interstitial invasive trophoblast cells move between blood vessels and directly infiltrate the uterine parenchyma [34,35,36]. In humans, invasive trophoblast cells are referred to as extravillous trophoblast (EVT) cells. Human EVT cells originate from a structure analogous to the junctional zone termed the EVT cell column [5,6,32] (Figure 2). Trophoblast giant cells are polyploid cells formed through a process termed endoreduplication [37,38]. Differentiation of spongiotrophoblast cells, glycogen trophoblast cells, and invasive trophoblast cells are less well understood. A connection between glycogen trophoblast cells and invasive trophoblast cells has been postulated [31] but not yet defined.
Placentation is a dynamic process characterized by significant changes in the structure and function of each placental compartment as gestation advances [13,39,40]. A few examples are noteworthy. Within the junctional zone, glycogen trophoblast cell and invasive trophoblast cell behaviors exhibit prominent temporal changes in numbers and distribution during the last half of pregnancy [13]. Glycogen trophoblast cells first appear after midgestation, exist in clusters, expand in numbers, and then disappear prior to parturition, whereas there is a prominent exit of invasive trophoblast cells from midgestation to term [34,35,36,41]. The appearance of trophoblast giant cells within the labyrinth zone is also gestation stage-dependent and a fixture during the latter days of pregnancy [41].
Many features of rat and mouse placentation are similar; however, prominent differences exist, including demarcation of the junctional and labyrinth compartments and especially the extent of intrauterine trophoblast cell invasion [12,13]. The boundary between placental compartments in the rat is precise resulting in a smooth interface, facilitating routine dissection, which is generally not possible for the mouse [39]. Trophoblast cell invasion in the mouse is shallow and underdeveloped relative to the extensive intrauterine infiltration observed for the rat [34].

3. Life Cycle of Invasive Trophoblast Cells

Deep intrauterine trophoblast cell invasion is a characteristic feature of rat placentation [12,13]. Invasive trophoblast cells possess the capacity to recognize, modify, and emulate the behaviors of cells within the uterus. Rat invasive trophoblast cells can be identified by their cytokeratin positive epithelial nature, expression of a unique subset of prolactin family genes and insulin-like growth factor II, accumulation of glycogen, and their position at the uterine site of placental attachment [13,20,34,35,36,42,43,44,45]. Invasive trophoblast cells originate in the junctional zone and migrate from the placenta and into the uterus [12,13] (Figure 3). The first wave of trophoblast cell invasion is restricted to endovascular trophoblast cell colonization of uterine spiral arteries/arterioles, which is initiated at the earliest stages of placentation and is a prominent feature of the midgestation mesometrial decidua [34,35,46]. Endovascular trophoblast cells mimic and supplant endothelial cells [34,35,47,48], destabilize vascular smooth muscle [49], and become embedded in fibrin [12]. A second wave of trophoblast cell invasion begins after gestation day 13.5, as endovascular trophoblast cells penetrate blood vessels beyond the decidua-myometrial boundary and interstitial trophoblast cell invasion is initiated [34,35,36,46]. By the end of pregnancy, interstitial invasive trophoblast cells have infiltrated the entire mesometrial compartment adjacent to the placenta, while some endovascular invasive trophoblast cells become flattened in appearance and embedded in the vascular wall associated with the reappearance of endothelium [34,35,36,46]. Expansion of invasive trophoblast cells within the uterine-placental interface is attributed to their derivation from progenitors located in the junctional zone and not their proliferation within the uterine-placental interface. Evidence exists for both diploid and polyploid invasive trophoblast cells [44,46]. The demise of intrauterine invasive trophoblast cells is signaled by parturition and removal of trophic factors emanating from the placenta [50]. Within a few days of delivery invasive trophoblast cells disappear, an event coupled to macrophage infiltration and restoration of vasomotor control of the uterine vascular smooth muscle [50]. The entire life cycle of rat invasive trophoblast cells transpires within a short interval of time. Invasive trophoblast cells can be tracked from their origin in the junctional zone, dissemination into the uterus, and demise [46,50]. Most importantly, these events exhibit elements of conservation with human placentation and are amenable to laboratory investigation.

4. Genetics of Trophoblast Cell Invasion

Strain-specific patterns of placentation are evident in the rat [51,52,53]. The Holtzman Sprague Dawley outbred and ACI, Fischer 344, and Dahl salt-sensitive inbred rat strains exhibit robust deep placentation, characterized by a well-developed junctional zone and widespread intrauterine trophoblast cell invasion [51,52,53]. In contrast, placentation in the Brown Norway rat is more restrained [51]. The most prominent features of the Brown Norway rat placentation site are a growth restricted placenta, including a thin junctional zone, and limited intrauterine trophoblast cell invasion [51,52,53,54]. Attenuated intrauterine trophoblast invasion is associated with retention of NK cells within the late gestation stage uterine-placental interface [51]. Pregnancy outcomes include placental insufficiency that negatively affects fetal growth and survival [51,54]. Brown Norway rat pregnancies are conspicuous for their small litter sizes [51,54]. Genetic influences on placental and fetal size are sexually dimorphic [54]. Brown Norway rat pregnancies exhibit a bias towards more pronounced male placental and female fetal growth restriction [54]. Potential sexual dimorphisms associated with intrauterine trophoblast cell invasion in the Brown Norway rat have not been reported. Chromosome substituted and recombinant inbred rat strains have been used to investigate chromosomal contributions to placental development [53,55]. In addition to structural differences, the Brown Norway rat placenta and especially the Brown Norway rat junctional zone possesses unique transcriptional profiles [53,54]. Among the affected transcripts was an upregulation of prolactin family 5, subfamily a, member 1 (Prl5a1) in the Brown Norway rat junctional zone [53]. Prl5a1 is an invasive trophoblast cell specific transcript, which is not typically expressed in the junctional zone [34,53]. Thus, providing evidence for an aberration in the exodus of invasive trophoblast cells from the Brown Norway rat junctional zone into the uterus. The Brown Norway rat invasive trophoblast cell phenotype is determined by intrinsic properties of trophoblast cells and an ineffectual maternal environment [52,53]. Although, gene regulatory pathways linked to abnormal Brown Norway rat trophoblast cell development have not been reported, insights into inadequacies associated with the Brown Norway rat maternal environment are apparent. Female Brown Norway rats are progesterone resistant and exhibit a compromised decidual response [52]. In humans, poor decidualization is an underlying cause of failed deep placentation resulting in disorders such as preeclampsia [56,57,58].

5. Experimental Manipulation of Trophoblast Cell Invasion and Uterine Spiral Artery Remodeling

Trophoblast cell-guided transformation of the uterus is influenced by environmental factors, the cellular constituents of the uterine-placental interface, an assortment of experimental manipulations, and disease states. Trophoblast cell invasion and uterine spiral artery remodeling can be influenced by targeting these structures within the uterus or through actions on the junctional zone and development of the invasive trophoblast cell lineage.

5.1. Oxygen Tension

Oxygen is an essential cellular nutrient and a critical regulator of hemochorial placentation [59,60]. Trophoblast cell development occurs in a low oxygen environment [59] and is under the control of hypoxia signaling pathways [60,61,62]. Exposure of pregnant rats to a hypoxic environment beginning after embryo implantation, and including gestation days 8.5 to 9.5, affects placental organization, resulting in an expansion of the junctional zone [46,63] and triggering enhanced development of endovascular invasive trophoblast cells, and their invasion, colonization, and restructuring of uterine spiral arteries [46]. Activation of the invasive trophoblast cell phenotype was guided by a hypoxia inducible factor (HIF) pathway involving HIF1B (also called aryl hydrocarbon nuclear translocator, (ARNT), lysine demethylase 3A (KDM3A), and matrix metallopeptidase 12 (MMP12) [64]. In response to low oxygen tension HIF activates the transcription of a cohort of genes, including KDM3A [47,64]. The trophoblast cell epigenetic landscape is redirected through the demethylating actions of KDM3A on histone 3 lysine 9 (H3K9), leading to further gene activation [64]. Among the genes activated in endovascular invasive trophoblast cells by this pathway is MMP12 [64,65]. Arterial elastin is degraded by MMP12 resulting in restructuring uterine spiral arteries [64,66,67]. The HIF-KDM3A-MMP12 pathway was elucidated using rat trophoblast stem (TS) cells and in vivo genetically manipulated rat models [64,68] and conservation of elements of the pathway determined using human placental tissue specimens and human trophoblast cells [64,65,66,67,69].
The relationship of oxygen and placentation is more complex than simply activating adaptations that facilitate deep placentation. Severe and/or chronic hypoxia and ischemia-reperfusion can lead to placental abnormalities, including disruptions in the junctional zone, impairments in intrauterine trophoblast cell invasion and uterine spiral artery remodeling, intrauterine fetal growth restriction, and failed pregnancy [70,71,72,73,74]. These adverse effects may be mediated via enothelin-1 signaling [70,74] and disruptions in matrix metallopeptidase expression [72]. Adverse consequences of poor oxygenation also emerge from failed placentation and compromised ability of the placenta to adapt to hypoxia [59,75,76]. Enhancing oxygen delivery can rescue some of the unfavorable effects of placental hypoxia [73].

5.2. Immune Cells

Two principal immune cell populations (NK cells, macrophages) have been linked to the regulation of trophoblast cell invasion and uterine spiral artery remodeling [77,78,79,80]. The uterine-placental interface exhibits dynamic changes in NK cell abundance, which shows a reciprocal relationship to the presence of invasive trophoblast cells [34,78,81]. NK cells are most prevalent after embryo implantation until midgestation and then diminish in number as gestation advances concomitant with increases in intrauterine trophoblast cell invasion [34,81]. The reciprocal relationship between NK cells and intrauterine trophoblast cell invasion is also demonstrated by the retention of NK cells within the uterine-placental interface in rat pregnancies with compromised intrauterine trophoblast cell invasion [48,51,82,83]. An exception is NK cell expansion in failed conceptus sites [84]. Macrophages are situated throughout the uterine-placental interface, including in locations proximal to uterine spiral arteries [85,86] and exhibit a pronounced increase in number at parturition [87,88,89].
In vivo depletion of NK cells using immunological or genetic approaches is an effective strategy for discerning the biology of NK cells during establishment of the uterine-placental interface [47,90]. Three key observations were made from pregnancies deficient in NK cells [47,90]: (i) NK cells contribute to early events in uterine spiral artery remodeling; (ii) NK cells restrain intrauterine endovascular trophoblast cell invasion; and (iii) placentation sites adapt in the absence of NK cells. NK cells act to disrupt the integrity of the tunica media of uterine arterial vessels decreasing vascular resistance and increasing blood flow to the placenta, engineering the first wave of uterine spiral artery remodeling [47,91]. Endovascular invasive trophoblast cells have similar actions on vascular smooth muscle, engineer a second wave of uterine spiral artery remodeling, and can compensate for the absence of NK cells [34,35,47,90]. The nature of NK cell restraint on endovascular trophoblast cell invasion may be linked to oxygen delivery [47]. NK cells restructure uterine spiral arteries to promote the transfer of oxygenated blood to the developing placenta and their absence leads to hypoxia-mediated adaptations within the placenta, including an expansion of the junctional zone (source of invasive trophoblast cells) and activation of endovascular trophoblast cell invasion into uterine spiral arteries [47,90]. It is also important to appreciate that NK cells are not uniform, they are heterogeneous, and their phenotype can shift from pro-pregnancy to embryotoxic depending on environmental signals and developmental outcomes [92,93,94].
Macrophages possess several roles at the uterine-placental interface [78,79,80]. They contribute to the regulation of angiogenesis, post-implantation uterine tissue repair, uterine spiral artery remodeling, and immune tolerance [79,80]. Most interestingly, they expand in number at term [87,88,89] and have been hypothesized to contribute to post-partum reconstruction of the uterus, including removal of invasive trophoblast cells [50,79,80,95,96]. Macrophages are also linked to pregnancy-dependent uterine-placental interface responses to inflammation and infection [97,98]. Lipopolysaccharide (LPS) exposure is an effective means of inducing an inflammatory response at the uterine-placental interface that is characterized by an increase in macrophage numbers and adverse effects on intrauterine trophoblast cell invasion and uterine spiral artery remodeling [97,99,100,101,102,103]. TLR4 activation and tumor necrosis factor alpha are viewed as drivers of the deleterious actions of LPS on the uterine-placental interface [97,104,105], whereas galectins and interleukin 33 can protect against the unfavorable consequences of LPS exposure [106,107]. Further implication of macrophages as modulators of the uterine-placental interface is achieved through their experimental activation via infusion of adenosine triphosphate (ATP) into pregnant rats [108]. ATP-induced macrophage activation results in restricted interstitial trophoblast cell invasion and impaired uterine spiral artery remodeling [108]. Maternal infection with specific strains of Porphyromonas gingivalis, a Gram-negative bacterium, affects the behavior of NK cells and macrophages and negatively impacts trophoblast cell-guided uterine spiral artery remodeling [98].

5.3. Drug, Toxicant, and Miscellaneous Exposures

There have been numerous investigations examining the effects of an assortment of drugs and toxicants on the uterine-placental interface with the goal of determining the target tissue mediating negative fetal developmental outcomes [40]. Treatment with chlorpromazine, a phenothiazine used as a tranquilizer, a range of anticancer drugs (cisplatin, 6-mercaptopurine, methyl methanesulfonate, and methotrexate), dexamethasone (glucocorticoid agonist), aryl hydrocarbon receptor agonists (β-naphthoflavone and Arolor 1254), GW501516 (peroxisome proliferator receptor agonist), nicotine, and thyroid hormone dysregulation cause reductions in placental size and structure, including hypoplasia of the junctional zone, abnormalities in junctional zone glycogen trophoblast cell development, and/or decreased intrauterine interstitial trophoblast cell invasion [109,110,111,112,113,114,115,116,117,118,119,120,121,122,123]. A potential relationship of junctional zone development and intrauterine trophoblast cell invasion is logical and informative. However, mechanisms underlying the effects of these various drugs on the uterine-placental interface are poorly understood.
Other agents act directly on the uterine-placental interface impacting the behavior of NK cells, invasive trophoblast cells, and/or uterine spiral artery remodeling. Exposure to tamoxifen, a selective estrogen receptor modulator, has little effect on placental morphogenesis but major disruptive effects on the uterine-placental interface, especially triggering decreases in uterine NK cell accumulation and defective uterine spiral artery transformation at midgestation [124]. In vivo treatment with doxycycline, a tetracycline antibiotic, impairs endovascular trophoblast cell invasion and uterine spiral artery remodeling and placental perfusion [82]. The mode of action of doxycycline may be through inhibition of matrix metallopeptidase activities [82]. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) also has profound effects on the uterine-placental interface, but in this case by accelerating endovascular trophoblast cell invasion [125]. Interestingly, in the rat the actions of TCDD are not directly on trophoblast cells [125]. Instead, TCDD signals through the aryl hydrocarbon receptor (AHR) in endothelial cells, which then act to modulate the cellular composition of the uterine-placental interface. β-naphthoflavone similarly targets endothelial cells of uterine spiral arteries but not trophoblast cells [123]. In addition to these conserved actions on endothelial cells, human trophoblast cells respond directly to activators of AHR signaling, a property not characteristic of rat trophoblast cells [125].
Maternal alcohol ingestion during pregnancy can lead to congenital anomalies, including placental and fetal growth restriction [126]. Timing and duration of ethanol exposure differentially affect placentation [127,128]. Chronic maternal ethanol intake during pregnancy in the rat adversely impacts placentation resulting in disruptions in the junctional zone [129,130,131], decreased prevalence of glycogen trophoblast cells, shallow intrauterine trophoblast cell invasion, and failure of uterine spiral artery remodeling [127,129,130]. Some of these aberrations in placentation may be connected to the disruptive effects of maternal ethanol exposure on insulin-like growth factor and NOTCH signaling [129,130]. In contrast, transient maternal ethanol intake around the time of conception leads to an expansion of the junctional zone and glycogen trophoblast cells [128]. The impact of this latter manipulation on intrauterine trophoblast cell invasion and uterine spiral artery remodeling was not reported.
Treatment with ketoconazole (antifungal drug) or a methylhydrazine derivative (anti-cancer drug) resulted in an expansion of the junctional zone, including glycogen trophoblast cell clusters [132,133]. Administration of phthalates, industrial plasticizers, to pregnant rats had contradictory actions [134]. Phthalate treatment increased placental size but led to degenerative changes within the junctional zone. The impact of ketoconazole, methylhydrazine, or phthalates on the invasive trophoblast cell lineage and uterine spiral artery remodeling was not reported.

5.4. Disease States

An assortment of disease states affecting pregnancy and placentation can be modeled in the rat. We will provide an overview of research specifically connecting health disorders that affect trophoblast cell-guided uterine spiral artery remodeling.

5.4.1. Diabetes

Diabetes during pregnancy has been simulated in the rat leading to maternal hyperglycemia with consequential effects on placental and fetal development [135]. Placentomegaly is a typical response to maternal hyperglycemia [136,137,138,139,140]. The junctional zone expands in size and glycogen trophoblast cell clusters become more abundant [136,137,138,139], which are characterized by prominent changes in the junctional zone transcriptome [140]. These events within the placentation site are connected to failures in intrauterine trophoblast cell invasion, especially the interstitial invasive trophoblast cell lineage [83,140], and impairments in uterine spiral artery remodeling [141]. Maternal hyperglycemia is also associated with a retention/expansion of NK cells and macrophages within the uterine-placental interface [83] and compromised plasticity of placentation to environmental stressors such as hypoxia [140]. Some of the consequences of maternal hyperglycemia may be mediated by the direct actions of elevated glucose on trophoblast cell development [140]. Mechanisms underlying the effects of maternal hyperglycemia on junctional zone expansion and its potential link to the invasive trophoblast cell lineage, and placental plasticity have not been determined.

5.4.2. Hypertension/Preeclampsia

Placental abnormalities are associated with pregnancy-induced hypertension and preeclampsia and can be the cause or consequence of these pregnancy-related diseases [10,142]. The literature is replete with descriptions of rat “models” of preeclampsia [143]. As a rule, the focus of the reports is generally on elevated maternal blood pressure and kidney dysfunction, which are hallmarks of the symptomology of the human clinical condition known as preeclampsia [143,144]. Prevailing evidence indicates that failures in trophoblast cell-guided uterine spiral artery are the underlying cause of early onset preeclampsia in humans [10,142]. Thus, there could be advantages in modeling these disorders in a species which exhibits deep placentation such as the rat [12,13]. Insults damaging development of the endovascular trophoblast cell lineage and endovascular trophoblast cell-mediated uterine spiral artery remodeling should be at the core of modeling preeclampsia. However, it is rare for research modeling preeclampsia in the rat to investigate biology at the placentation site, especially as the cause of the disorder. It is also important to appreciate a disease process such as preeclampsia is inherently problematic to investigate in a polytocous species such as the rat. Litter size and intrauterine position can influence fetal development [145,146,147,148]. The intrinsic response of a litter-bearing species to an insult that compromises maternal nutrient delivery to the fetus is to sacrifice the health of vulnerable conceptuses allowing the fittest to survive. Experimentally, this becomes a challenge since it is common to observe a range of placental and fetal responses to an insult within the same pregnancy creating an inherent bias for the analysis. In this section the goal is to highlight experimental manipulations causing maternal hypertension or a “preeclampsia-like” condition and their effects on trophoblast cell-guided uterine spiral artery remodeling. In general, these represent the consequences of induction of maternal hypertension or the “preeclampsia-like” condition rather than being the cause of the disorder.
A frequently studied transgenic rat model possessing an activated renin-angiotensin system exhibits maternal hypertension and many of the hallmarks of preeclampsia [149,150,151]. Maternal hypertension arises from mating female rats expressing an angiotensinogen transgene with male rats expressing a human renin transgene [149]. In contrast to predictions based on preeclampsia in humans, this model of preeclampsia displayed enhanced endovascular trophoblast cell invasion and uterine spiral artery remodeling [152,153,154]. Differences in endovascular trophoblast cell invasion were not as pronounced by the end of gestation [153]. The findings could be interpreted to reflect plasticity, a healthy placentation site, and a predictable response to poor perfusion and deprived oxygen delivery during critical early stages of placental morphogenesis [46]. Interestingly, target tissue access to angiotensin II is relevant to the biological outcome observed with this transgenic model. Uteroplacental angiotensin II upregulation correlates with endovascular trophoblast cell invasion and uterine spiral artery remodeling, whereas systemic angiotensin II delivery inhibits trophoblast cell invasion and vascular remodeling [154]. NK cells provide a protective role in this maternal hypertension model [155]. Immune depletion of NK cells after the initiation of intrauterine trophoblast cell invasion (gestation day 15) specifically inhibited interstitial trophoblast cell invasion, but not endovascular trophoblast cell invasion, and resulted in uterine vasculopathy [155].
The stroke prone spontaneous hypertensive rat (SHRSP) has also been used as a model for pregnancy research. The SHRSP model was established through phenotypic selection and inbreeding [156]. Unlike the transgenic model described above, hypertension in the SHRSP rat is not restricted to pregnancy [157]. SHRSP pregnancies are characterized by small litter sizes and placental and fetal growth restriction [157]. Placental growth restriction includes an underdeveloped junctional zone and diminished glycogen trophoblast cells. The SHRSP uterine-placental interface possesses decreased trophoblast cell invasion and vascular remodeling and unlike other examples of shallow trophoblast cell invasion, NK cells do not compensate and expand in number but instead are diminished [157]. The selection process and multigenic phenotype characteristic of the SHRSP model makes analysis a challenge, including the identification of suitable controls for experiments.
A surgical model causing reduced uterine perfusion pressure (RUPP) during the last week of pregnancy has been utilized to model aspects of preeclampsia in the rat [143,158]. As noted by others, the RUPP rat is useful for modeling the consequences of compromised blood delivery to the placenta and thus several aspects of preeclampsia but is not appropriate for investigating a placental etiology for the disorder [158]. The RUPP procedure leads to an expanded junctional zone [159] and impairments in both intrauterine trophoblast invasion and uterine spiral artery remodeling [72].
Some other attempts to model maternal hypertension/preeclampsia in the rat and examine the consequences on intrauterine trophoblast cell guided uterine spiral artery remodeling include maternal hyperinsulinemia, inhibition of heme oxygenase (HO), and treatment with growth arrest-specific 6 (GAS6) [160,161,162,163,164]. All manipulations resulted in elevated blood pressure and intrauterine growth restriction but caused a range of phenotypes at the uterine-placental interface. Endovascular invasive trophoblast cells were more abundant and located deeper within the uterine parenchyma in pregnancies with maternal hyperinsulinemia [161], whereas HO inhibition led to diminished endovascular trophoblast cell-guided uterine spiral artery remodeling [163]. In contrast, endovascular invasive trophoblast cells were not a target of exogenous GAS6 injections but instead, intrauterine interstitial trophoblast cell invasion was diminished [164]. The range of phenotypic responses to these experimental manipulations reflect the complexity of regulatory events controlling intrauterine trophoblast cell invasion and uterine vascular remodeling.

5.4.3. Malnutrition/Obesity/Hyperthermia

The rat uterine-placental interface has been examined following manipulations of diet and other pathophysiologic challenges. Again, our attention focuses on responses of the junctional zone and events at the uterine-placental interface. Generalized maternal undernutrition, protein restriction, and hyperthermia negatively impacted growth of the junctional zone [165,166,167]. In contrast, maternal iron deficiency resulted in an expansion of the size of the junctional zone [168]. Whether these manipulations affect development of the invasive trophoblast cell lineage and/or trophoblast-guided uterine spiral artery remodeling was not reported. A high fat diet results in a diminished junctional zone [169], an early increase in endovascular and interstitial trophoblast cell invasion and later decreased interstitial trophoblast cell invasion [170].

5.4.4. Overview

To a large extent, the preceding sections on the regulation of invasive trophoblast cell-guided uterine spiral artery remodeling, which are summarized in Table 1, have described potentially interesting phenomenology but provide limited insight into mechanisms regulating the uterine-placental interface.

6. A Path to Conserved Mechanisms Controlling Invasive Trophoblast Cell-Guided Uterine Transformation

As detailed above, the rat has limitations as a model for investigating human pregnancy-related diseases. However, the rat can be utilized to investigate specific stages of invasive trophoblast cell lineage development and uterine spiral artery remodeling [12,13]. These stages include: (i) cellular dynamics within the junctional zone, a structure analogous to the EVT cell column of the human placentation site, representing respective sites where the invasive trophoblast cell lineage originates; (ii) differentiation of interstitial and endovascular invasive trophoblast cells, and (iii) actions of invasive trophoblast cells on the uterine parenchyma and vasculature. Initially, candidate conserved gene regulatory networks are identified by gene/protein expression profiling in rat and human placentation. Cellular mechanisms can be interrogated using in vitro rat trophoblast stem cell models, including the Rcho-1 trophoblast stem cell line [171] and blastocyst-derived trophoblast stem cell lines [68] and conservation determined using human trophoblast stem cells [172]. It is important to appreciate that all in vitro analyses are artificial and potentially misleading without complementary in vivo physiological assessments [5]. Invasive trophoblast cells can be effectively tracked in vivo [46,173] and the importance of a gene in a physiological context can be evaluated using lentiviral-mediated trophectoderm gene manipulation [174,175] and global genome edited rat models [48,64,125,176]. Roles for phosphoinositide 3-kinase (PI3K)/AKT/fos-related antigen 1 (FOSL1)/JUNB proto-oncogene (JUNB), HIF/KDM3A/MMP12, achaete-scute family bHLH transcription factor 2 (ASCL2), and tissue factor pathway inhibitor (TFPI) as conserved regulators of the invasive trophoblast cell lineage have been demonstrated using this strategy [47,48,64,176,177,178,179,180] (Table 2). PI3K/AKT signaling regulates FOSL1, which cooperates with JUNB, to form a heterodimer and a functional activator protein 1 (AP-1) transcription factor that facilitates endovascular trophoblast cell development during early stages of placentation [177,178,179,180]. As discussed above, the HIF/KDM3A/MMP12 regulatory circuit contributes to invasive trophoblast cell lineage decisions in response to hypoxia [47,64]. ASCL2, a transcription factor, and TFPI, a regulator of hemostasis, act independently to promote invasive trophoblast cell differentiation [48,176]. Results from the experimentation establish the feasibility of the outlined research strategy and identify key nodes of regulation impacting the invasive trophoblast cell lineage and trophoblast-guided uterine spiral artery remodeling.

7. Final Thoughts

The placenta is an organ essential to viviparity [181]. Most importantly, maternal resources are redirected through the actions of the placenta to ensure fetal development. As described above, among mammals this task is accomplished using different strategies [3,11,181]. In species with deep placentation, such as the human and rat, trophoblast cells penetrate deep into the uterus to reengineer maternal vasculature, which supply nutrients to the placenta and for transfer to the fetus [4,6,11]. This is a remarkable undertaking. Trophoblast stem/progenitor cells differentiate into invasive trophoblast cells, become motile and capable of traversing and restructuring the uterus, and modulating uterine cell function [5,6]. These tasks must be navigated within the challenges of a genetically foreign landscape and a potentially volatile maternal environment. Invasive trophoblast cells evolved to facilitate viviparity. At this juncture, our knowledge of invasive trophoblast cells is largely descriptive. Tools are needed, especially tractable in vivo model systems, and the implementation of in vivo imaging platforms to explore trophoblast-guided uterine spiral artery remodeling and its impact on oxygen and nutrient delivery. Questions abound regarding the biology of invasive trophoblast cell transformation of the uterus: (i) How do we identify trophoblast stem/progenitor cells that are destined to be invasive trophoblast cells and where are they located within the placenta? In human placentation, these cells are situated at the base of the EVT cell column [6], but this is not the case for the junctional zone of the rat placenta. (ii) What are the regulatory events controlling the expansion, commitment, and differentiation of trophoblast stem/progenitor cells to the invasive trophoblast cell lineage? (iii) What dictates differentiation to an endovascular invasive trophoblast cell versus an interstitial invasive trophoblast cell? These differentiation outcomes may be positional and determined by the proximal milieu or set-up by the systematic activation or inactivation of specific regulatory pathways. (iv) What mechanisms do endovascular and interstitial invasive trophoblast cells use to exit the placenta and traverse the uterus? Cell guidance systems within a blood vessel are different than present in the decidual/stromal compartment. (v) What is the biological importance of endovascular versus interstitial invasive trophoblast cells? (vi) What is the nature of the invasive trophoblast cell-immune cell dialog? Invasive trophoblast cells and NK cells possess similar uterine targets, but their actions are temporally and physically separated. Differences are apparent regarding the relationships of endovascular versus interstitial invasive trophoblast cells and NK cells and possibly macrophages. Invasive trophoblast cell properties ensuring immune protection are not well understood. (vii) How does the uterus control the life cycle of invasive trophoblast cells? (viii) What regulates the demise of invasive trophoblast cells from the uterus following parturition? The rat has much to offer as an in vivo model for investigating invasive trophoblast cells and their biological functions. It is important to appreciate that not all regulatory pathways controlling the uterine-placental interface will be conserved in the rat and human. Species specific regulatory pathways provide insights into nodes of specialization required to address unique needs for a species. Further development and implementation of tools to examine and manipulate individual cell lineages within the uterine-placental are essential for future progress.

Author Contributions

Conceptualization, writing, and original draft preparation, V.S. and M.J.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the University of Kansas Medical Center Biomedical Training Program (VS), Kansas Idea Network of Biomedical Research Excellence, P20 GM103418 (VS), NIH grants (HD020676, HD079363, HD099638, ES029280), and the Sosland Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Supported by the University of Kansas Medical Center Biomedical Training Program (VS), Kansas Idea Network of Biomedical Research Excellence, P20 GM103418 (VS), NIH grants (HD020676, HD079363, HD099638, ES029280), and the Sosland Foundation. We thank Stacy Oxley and Brandi Miller for administrative assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Burton, G.J.; Fowden, A.L.; Thornburg, K.L. Placental origins of chronic disease. Physiol. Rev. 2016, 96, 1509–1565. [Google Scholar] [CrossRef] [PubMed]
  2. Maltepe, E.; Fisher, S.J. Placenta: The forgotten organ. Annu. Rev. Cell. Dev. Biol. 2015, 31, 523–552. [Google Scholar] [CrossRef] [PubMed]
  3. Roberts, R.M.; Green, J.A.; Schulz, L.C. The evolution of the placenta. Reproduction 2016, 152, R179–R189. [Google Scholar] [CrossRef] [Green Version]
  4. Pijnenborg, R.; Robertson, W.B.; Brosens, I.; Dixon, G. Trophoblast invasion and the establishment of haemochorial placentation in man and laboratory animals. Placenta 1981, 2, 71–91. [Google Scholar] [CrossRef]
  5. Soares, M.J.; Varberg, K.M.; Iqbal, K. Hemochorial placentation: Development, function, and adaptations. Biol. Reprod. 2018, 99, 196–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Knöfler, M.; Haider, S.; Saleh, L.; Pollheimer, J.; Gamage, T.K.J.B.; James, J. Human placenta and trophoblast development: Key molecular mechanisms and model systems. Cell Mol. Life Sci. 2019, 76, 3479–3496. [Google Scholar] [CrossRef] [Green Version]
  7. Pijnenborg, R.; Vercruysse, L.; Hanssens, M. The uterine spiral arteries in human pregnancy: Facts and controversies. Placenta 2006, 27, 939–958. [Google Scholar] [CrossRef]
  8. Pollheimer, J.; Vondra, S.; Baltayeva, J.; Beristain, A.G.; Knöfler, M. Regulation of placental extravillous trophoblasts by the maternal uterine environment. Front. Immunol. 2018, 9, 2597. [Google Scholar] [CrossRef]
  9. 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. [Google Scholar] [CrossRef] [Green Version]
  10. Brosens, I.; Puttemans, P.; Benagiano, G. Placental bed research: I. The placental bed: From spiral arteries remodeling to the great obstetrical syndromes. Am. J. Obstet. Gynecol. 2019, 221, 437–456. [Google Scholar] [CrossRef]
  11. Wooding, P.; Burton, G. Comparative Placentation; Springer: Heidelberg, Germany, 2008. [Google Scholar]
  12. Pijnenborg, R.; Vercruysse, L. Animal models of deep trophoblast invasion. In Placental Bed Disorders; Pijnenborg, R., Brosens, I., Romero, R., Eds.; Cambridge University Press: Cambridge, UK, 2010; pp. 127–139. [Google Scholar]
  13. Soares, M.J.; Chakraborty, D.; Rumi, M.A.K.; Konno, T.; Renaud, S.J. Rat placentation: An experimental model for investigating the hemochorial maternal-fetal interface. Placenta 2012, 33, 233–243. [Google Scholar] [CrossRef] [Green Version]
  14. Cha, J.; Sun, X.; Dey, S.K. Mechanisms of implantation: Strategies for successful pregnancy. Nat. Med. 2012, 18, 1754–1767. [Google Scholar] [CrossRef] [PubMed]
  15. Erlebacher, A. Immunology of the maternal-fetal interface. Annu. Rev. Immunol. 2013, 31, 387–411. [Google Scholar] [CrossRef] [PubMed]
  16. Turco, M.Y.; Moffett, A. Development of the human placenta. Development 2019, 146, dev163428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Bridgman, J. A morphological study of the development of the placenta of the rat; an histological and cytological study of the development of the chorioallantoic placenta of the white rat. J. Morphol. 1948, 83, 195–223. [Google Scholar] [CrossRef]
  18. Enders, A.C. A comparative study of the fine structure of the trophoblast in several hemochorial placentas. Am. J. Anat. 1965, 116, 29–67. [Google Scholar] [CrossRef]
  19. Jollie, W.P. Fine structural changes in the junctional zone of the rat placenta with increasing gestational age. J. Ultrastruct. Res. 1965, 12, 420–438. [Google Scholar] [CrossRef]
  20. Davies, J.; Glasser, S.R. Histological and fine structural observations on the placenta of the rat. Acta Anat. 1968, 69, 542–608. [Google Scholar] [CrossRef]
  21. Rossant, J. Stem cells from the mammalian blastocyst. Stem Cells 2001, 19, 477–482. [Google Scholar] [CrossRef]
  22. Gardner, R.L.; Beddington, R.S. Multi-lineage ‘stem’ cells in the mammalian embryo. J. Cell Sci. Suppl. 1988, 10, 11–27. [Google Scholar] [CrossRef] [Green Version]
  23. Simmons, D.G.; Cross, J.C. Determinants of trophoblast lineage and cell subtype specification in the mouse placenta. Dev. Biol. 2005, 284, 12–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Walentin, K.; Hinze, C.; Schmidt-Ott, K.M. The basal chorionic trophoblast cell layer: An emerging coordinator of placenta development. Bioessays 2016, 38, 254–265. [Google Scholar] [CrossRef] [PubMed]
  25. Soares, M.J.; Chapman, B.M.; Rasmussen, C.A.; Dai, G.; Kamei, T.; Orwig, K.E. Differentiation of trophoblast endocrine cells. Placenta 1996, 17, 277–289. [Google Scholar] [CrossRef]
  26. Cross, J.C.; Nakano, H.; Natale, D.R.; Simmons, D.G.; Watson, E.D. Branching morphogenesis during development of placental villi. Differentiation 2006, 74, 393–401. [Google Scholar] [CrossRef] [PubMed]
  27. Knipp, G.T.; Audus, K.L.; Soares, M.J. Nutrient transport across the placenta. Adv. Drug Deliv. Rev. 1999, 38, 41–58. [Google Scholar] [CrossRef]
  28. Watson, E.D.; Cross, J.C. Development of structures and transport functions in the mouse placenta. Physiology 2005, 20, 180–193. [Google Scholar] [CrossRef] [PubMed]
  29. Peel, S.; Bulmer, D. Proliferation and differentiation of trophoblast in the establishment of the rat chorio-allantoic placenta. J. Anat. 1977, 124 Pt 3, 675–687. [Google Scholar]
  30. John, R.M. Epigenetic regulation of placental endocrine lineages and complications of pregnancy. Biochem. Soc. Trans. 2013, 41, 701–709. [Google Scholar] [CrossRef] [Green Version]
  31. Tunster, S.J.; Watson, E.D.; Fowden, A.L.; Burton, G.J. Placental glycogen stores and fetal growth: Insights from genetic mouse models. Reproduction 2020, 159, R213–R235. [Google Scholar] [CrossRef]
  32. Varberg, K.M.; Soares, M.J. Paradigms for investigating invasive trophoblast cell development and contributions to uterine spiral artery remodeling. Placenta 2021, 113, 48–56. [Google Scholar] [CrossRef]
  33. Selye, H.; McKeown, T. Studies on the physiology of the maternal placenta in the rat. Proc. R. Soc. Lond. Biol. 1935, 119, 1–31. [Google Scholar]
  34. Ain, R.; Canham, L.N.; Soares, M.J. Gestation stage-dependent intrauterine trophoblast cell invasion in the rat and mouse: Novel endocrine phenotype and regulation. Dev. Biol. 2003, 260, 176–190. [Google Scholar] [CrossRef] [Green Version]
  35. Caluwaerts, S.; Vercruysse, L.; Luyten, C.; Pijnenborg, R. Endovascular trophoblast invasion and associated structural changes in uterine spiral arteries of the pregnant rat. Placenta 2005, 26, 574–584. [Google Scholar] [CrossRef] [PubMed]
  36. Vercruysse, L.; Caluwaerts, S.; Luyten, C.; Pijnenborg, R. Interstitial trophoblast invasion in the decidua and mesometrial triangle during the last third of pregnancy in the rat. Placenta 2006, 27, 22–33. [Google Scholar] [CrossRef] [PubMed]
  37. Hu, D.; Cross, J.C. Development and function of trophoblast giant cells in the rodent placenta. Int. J. Dev. Biol. 2010, 54, 341–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Zybina, T.G.; Zybina, E.V. Role of cell cycling and polyploidy in placental trophoblast of different mammalian species. Reprod. Domest. Anim. 2020, 55, 895–904. [Google Scholar] [CrossRef] [PubMed]
  39. Ain, R.; Konno, T.; Canham, L.N.; Soares, M.J. Phenotypic analysis of the rat placenta. Methods Mol. Med. 2006, 121, 295–313. [Google Scholar]
  40. Furukawa, S.; Tsuji, N.; Sugiyama, A. Morphology and physiology of rat placenta for toxicological evaluation. J. Toxicol. Pathol. 2019, 32, 1–17. [Google Scholar] [CrossRef] [Green Version]
  41. Campbell, W.J.; Deb, S.; Kwok, S.C.; Joslin, J.A.; Soares, M.J. Differential expression of placental lactogen-II and prolactin-like protein-A in the rat chorioallantoic placenta. Endocrinology 1989, 125, 1565–1574. [Google Scholar] [CrossRef]
  42. Correia-da-Silva, G.; Bell, S.C.; Pringle, J.H.; Teixeira, N. Expression of mRNA encoding insulin-like growth factors I and II by uterine tissues and placenta during pregnancy in the rat. Mol. Reprod. Dev. 1999, 53, 294–305. [Google Scholar] [CrossRef]
  43. Toft, D.J.; Linzer, D.I. Identification of three prolactin-related hormones as markers of invasive trophoblasts in the rat. Biol. Reprod. 2000, 63, 519–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zybina, T.G.; Stein, G.I.; Zybina, E.V. Endopolyploid and proliferating trophoblast cells express different patterns of intracellular cytokeratin and glycogen localization in the rat placenta. Cell Biol. Int. 2011, 35, 649–655. [Google Scholar] [CrossRef] [PubMed]
  45. Wiemers, D.O.; Ain, R.; Ohboshi, S.; Soares, M.J. Migratory trophoblast cells express a newly identified member of the prolactin gene family. J. Endocrinol. 2003, 179, 335–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Rosario, G.X.; Konno, T.; Soares, M.J. Maternal hypoxia activates endovascular trophoblast cell invasion. Dev. Biol. 2008, 314, 362–375. [Google Scholar] [CrossRef] [Green Version]
  47. Chakraborty, D.; Rumi, M.A.; Konno, T.; Soares, M.J. Natural killer cells direct hemochorial placentation by regulating hypoxia-inducible factor dependent trophoblast lineage decisions. Proc. Natl. Acad. Sci. USA 2011, 108, 16295–16300. [Google Scholar] [CrossRef] [Green Version]
  48. Muto, M.; Chakraborty, D.; Varberg, K.M.; Moreno-Irusta, A.; Iqbal, K.; Scott, R.L.; McNally, R.P.; Choudhury, R.H.; Aplin, J.D.; Okae, H.; et al. Intersection of regulatory pathways controlling hemostasis and hemochorial placentation. Proc. Natl. Acad. Sci. USA 2021, 118, e2111267118. [Google Scholar] [CrossRef]
  49. Nandy, D.; Das, S.; Islam, S.; Ain, R. Molecular regulation of vascular smooth muscle cell phenotype switching by trophoblast cells at the maternal-fetal interface. Placenta 2020, 93, 64–73. [Google Scholar] [CrossRef]
  50. Rosario, G.X.; Ain, R.; Konno, T.; Soares, M.J. Intrauterine fate of invasive trophoblast cells. Placenta 2009, 30, 457–463. [Google Scholar] [CrossRef] [Green Version]
  51. Konno, T.; Rempel, L.A.; Arroyo, J.A.; Soares, M.J. Pregnancy in the brown Norway rat: A model for investigating the genetics of placentation. Biol. Reprod. 2007, 76, 709–718. [Google Scholar] [CrossRef] [Green Version]
  52. Konno, T.; Graham, A.R.; Rempel, L.A.; Ho-Chen, J.K.; Alam, S.M.; Bu, P.; Rumi, M.A.; Soares, M.J. Subfertility linked to combined luteal insufficiency and uterine progesterone resistance. Endocrinology 2010, 151, 4537–4550. [Google Scholar] [CrossRef] [Green Version]
  53. Konno, T.; Rempel, L.A.; Rumi, M.A.; Graham, A.R.; Asanoma, K.; Renaud, S.J.; Soares, M.J. Chromosome-substituted rat strains provide insights into the genetics of placentation. Physiol. Genom. 2011, 43, 930–941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Goyal, R.; Yellon, S.M.; Longo, L.D.; Mata-Greenwood, E. Placental gene expression in a rat ‘model’ of placental insufficiency. Placenta 2010, 31, 568–575. [Google Scholar] [CrossRef] [PubMed]
  55. Buresova, M.; Zidek, V.; Musilova, A.; Simakova, M.; Fucikova, A.; Bila, V.; Kren, V.; Kazdova, L.; Di Nicolantonio, R.; Pravenec, M. Genetic relationship between placental and fetal weights and markers of the metabolic syndrome in rat recombinant inbred strains. Physiol. Genom. 2006, 26, 226–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Conrad, K.P.; Rabaglino, M.B.; Post Uiterweer, E.D. Emerging role for dysregulated decidualization in the genesis of preeclampsia. Placenta 2017, 60, 119–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Garrido-Gomez, T.; Dominguez, F.; Quiñonero, A.; Diaz-Gimeno, P.; Kapidzic, M.; Gormley, M.; Ona, K.; Padilla-Iserte, P.; McMaster, M.; Genbacev, O.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Garrido-Gómez, T.; Castillo-Marco, N.; Cordero, T.; Simón, C. Decidualization resistance in the origin of preeclampsia. Am. J. Obstet. Gynecol. 2020, 226, S886–S894. [Google Scholar] [CrossRef] [PubMed]
  59. Burton, G.J. Oxygen, the Janus gas; its effects on human placental development and function. J. Anat. 2009, 215, 27–35. [Google Scholar] [CrossRef] [Green Version]
  60. Fryer, B.H.; Simon, M.C. Hypoxia, HIF and the placenta. Cell Cycle 2006, 5, 495–498. [Google Scholar] [CrossRef]
  61. Soares, M.J.; Iqbal, K.; Kozai, K. Hypoxia and placental development. Birth Defects Res. 2017, 109, 1309–1329. [Google Scholar] [CrossRef]
  62. Chang, C.W.; Wakeland, A.K.; Parast, M.M. Trophoblast lineage specification, differentiation and their regulation by oxygen tension. J. Endocrinol. 2018, 236, R43–R56. [Google Scholar] [CrossRef] [Green Version]
  63. Nuzzo, A.M.; Camm, E.J.; Sferruzzi-Perri, A.N.; Ashmore, T.J.; Yung, H.W.; Cindrova-Davies, T.; Spiroski, A.M.; Sutherland, M.R.; Logan, A.; Austin-Williams, S.; et al. Placental adaptation to early-onset hypoxic pregnancy and mitochondria-targeted antioxidant therapy in a rodent model. Am. J. Pathol. 2018, 188, 2704–2716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chakraborty, D.; Cui, W.; Rosario, G.X.; Scott, R.L.; Dhakal, P.; Renaud, S.J.; Tachibana, M.; Rumi, M.A.; Mason, C.W.; Krieg, A.J.; et al. HIF-KDM3A-MMP12 regulatory circuit ensures trophoblast plasticity and placental adaptations to hypoxia. Proc. Natl. Acad. Sci. USA 2016, 113, E7212–E7221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Hiden, U.; Eyth, C.P.; Majali-Martinez, A.; Desoye, G.; Tam-Amersdorfer, C.; Huppertz, B.; Ghaffari Tabrizi-Wizsy, N. Expression of matrix metallopeptidase 12 is highly specific for non-proliferating invasive trophoblasts in the first trimester and temporally regulated by oxygen-dependent mechanisms including HIF-1A. Histochem. Cell Biol. 2018, 149, 31–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Harris, L.K.; Keogh, R.J.; Wareing, M.; Baker, P.N.; Cartwright, J.E.; Whitley, G.S.; Aplin, J.D. BeWo cells stimulate smooth muscle cell apoptosis and elastin breakdown in a model of spiral artery transformation. Hum. Reprod. 2007, 22, 2834–2841. [Google Scholar] [CrossRef] [Green Version]
  67. Harris, L.K.; Smith, S.D.; Keogh, R.J.; Jones, R.L.; Baker, P.N.; Knöfler, M.; Cartwright, J.E.; Whitley, G.S.; Aplin, J.D. Trophoblast- and vascular smooth muscle cell-derived MMP-12 mediates elastolysis during uterine spiral artery remodeling. Am. J. Pathol. 2010, 177, 2103–2115. [Google Scholar] [CrossRef]
  68. Asanoma, K.; Rumi, M.A.; Kent, L.N.; Chakraborty, D.; Renaud, S.J.; Wake, N.; Lee, D.S.; Kubota, K.; Soares, M.J. FGF4-dependent stem cells derived from rat blastocysts differentiate along the trophoblast lineage. Dev. Biol. 2011, 351, 110–119. [Google Scholar] [CrossRef] [Green Version]
  69. Wakeland, A.K.; Soncin, F.; Moretto-Zita, M.; Chang, C.W.; Horii, M.; Pizzo, D.; Nelson, K.K.; Laurent, L.C.; Parast, M.M. Hypoxia directs human extravillous trophoblast differentiation in a hypoxia-inducible factor-dependent manner. Am. J. Pathol. 2017, 187, 767–780. [Google Scholar] [CrossRef] [Green Version]
  70. Zhou, J.; Xiao, D.; Hu, Y.; Wang, Z.; Paradis, A.; Mata-Greenwood, E.; Zhang, L. Gestational hypoxia induces preeclampsia-like symptoms via heightened endothelin-1 signaling in pregnant rats. Hypertension 2013, 62, 599–607. [Google Scholar] [CrossRef] [Green Version]
  71. Kimball, R.; Wayment, M.; Merrill, D.; Wahlquist, T.; Reynolds, P.R.; Arroyo, J.A. Hypoxia reduces placental mTOR activation in a hypoxia-induced model of intrauterine growth restriction (IUGR). Physiol. Rep. 2015, 3, e12651. [Google Scholar] [CrossRef]
  72. Lin, C.; He, H.; Cui, N.; Ren, Z.; Zhu, M.; Khalil, R.A. Decreased uterine vascularization and uterine arterial expansive remodeling with reduced matrix metalloproteinase-2 and -9 in hypertensive pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H165–H180. [Google Scholar] [CrossRef]
  73. Li, H.; Ohta, H.; Tahara, Y.; Nakamura, S.; Taguchi, K.; Nakagawa, M.; Oishi, Y.; Goto, Y.; Wada, K.; Kaga, M.; et al. Artificial oxygen carriers rescue placental hypoxia and improve fetal development in the rat pre-eclampsia model. Sci. Rep. 2015, 5, 15271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Thaete, L.G.; Khan, S.; Neerhof, M.G. Endothelin receptor A antagonism prevents damage to glycogen-rich placental cells following uterine ischemia-reperfusion in the rat. Reprod. Sci. 2016, 23, 1518–1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Zhang, D.; Liu, H.; Zeng, J.; Miao, X.; Huang, W.; Chen, H.; Huang, Y.; Li, Y.; Ye, D. Glucocorticoid exposure in early placentation induces preeclampsia in rats via interfering trophoblast development. Gen. Comp. Endocrinol. 2016, 225, 61–70. [Google Scholar] [CrossRef] [PubMed]
  76. Soares, M.J.; Chakraborty, D.; Kubota, K.; Renaud, S.J.; Rumi, M.A. Adaptive mechanisms controlling uterine spiral artery remodeling during the establishment of pregnancy. Int. J. Dev. Biol. 2014, 58, 247–259. [Google Scholar] [CrossRef] [Green Version]
  77. Chakraborty, D.; Rumi, M.A.; Soares, M.J. NK cells, hypoxia and trophoblast cell differentiation. Cell Cycle 2012, 11, 2427–2430. [Google Scholar] [CrossRef] [Green Version]
  78. Faas, M.M.; de Vos, P. Uterine NK cells and macrophages in pregnancy. Placenta 2017, 56, 44–52. [Google Scholar] [CrossRef]
  79. Jena, M.K.; Nayak, N.; Chen, K.; Nayak, N.R. Role of macrophages in pregnancy and related complications. Arch. Immunol. Ther. Exp. 2019, 67, 295–309. [Google Scholar] [CrossRef]
  80. Mor, G.; Abrahams, V.M. Potential role of macrophages as immunoregulators of pregnancy. Reprod. Biol. Endocrinol. 2003, 1, 119. [Google Scholar] [CrossRef] [Green Version]
  81. Head, J.R. Uterine natural killer cells during pregnancy in rodents. Nat. Immun. 1996, 15, 7–21. [Google Scholar]
  82. Verlohren, S.; Geusens, N.; Morton, J.; Verhaegen, I.; Hering, L.; Herse, F.; Dudenhausen, J.W.; Muller, D.N.; Luft, F.C.; Cartwright, J.E.; et al. Inhibition of trophoblast-induced spiral artery remodeling reduces placental perfusion in rat pregnancy. Hypertension 2010, 56, 304–310. [Google Scholar] [CrossRef] [Green Version]
  83. Groen, B.; Uuldriks, G.A.; de Vos, P.; Visser, J.T.; Links, T.P.; Faas, M.M. Impaired trophoblast invasion and increased numbers of immune cells at day 18 of pregnancy in the mesometrial triangle of type 1 diabetic rats. Placenta 2015, 36, 142–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Fonseca, B.M.; Almada, M.; Costa, M.A.; Teixeira, N.A.; Correia-da-Silva, G. Rat spontaneous foetal resorption: Altered α2-macroglobulin levels and uNK cell number. Histochem. Cell Biol. 2014, 142, 693–701. [Google Scholar] [CrossRef] [PubMed]
  85. Melgert, B.N.; Spaans, F.; Borghuis, T.; Klok, P.A.; Groen, B.; Bolt, A.; de Vos, P.; van Pampus, M.G.; Wong, T.Y.; van Goor, H.; et al. Pregnancy and preeclampsia affect monocyte subsets in humans and rats. PLoS ONE 2012, 7, e45229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Tessier, D.R.; Raha, S.; Holloway, A.C.; Yockell-Lelièvre, J.; Tayade, C.; Gruslin, A. Characterization of immune cells and cytokine localization in the rat utero-placental unit mid- to late gestation. J. Reprod. Immunol. 2015, 110, 89–101. [Google Scholar] [CrossRef]
  87. Brandes, D.; Anton, E. An electron microscopic cytochemical study of macrophages during uterine involution. J. Cell Biol. 1969, 41, 450–461. [Google Scholar] [CrossRef]
  88. Padykula, H.A.; Tansey, T.R. The occurrence of uterine stromal and intraepithelial monocytes and heterophils during normal late pregnancy in the rat. Anat. Rec. 1979, 193, 329–356. [Google Scholar] [CrossRef]
  89. Hamilton, S.; Oomomian, Y.; Stephen, G.; Shynlova, O.; Tower, C.L.; Garrod, A.; Lye, S.J.; Jones, R.L. Macrophages infiltrate the human and rat decidua during term and preterm labor: Evidence that decidual inflammation precedes labor. Biol. Reprod. 2012, 86, 39. [Google Scholar] [CrossRef]
  90. Renaud, S.J.; Scott, R.L.; Chakraborty, D.; Rumi, M.A.; Soares, M.J. Natural killer-cell deficiency alters placental development in rats. Biol. Reprod. 2017, 96, 145–158. [Google Scholar] [CrossRef] [Green Version]
  91. Zhang, J.; Chen, Z.; Smith, G.N.; Croy, B.A. Natural killer cell-triggered vascular transformation: Maternal care before birth? Cell. Mol. Immunol. 2011, 8, 1–11. [Google Scholar] [CrossRef] [Green Version]
  92. Hatta, K.; MacLeod, R.J.; Gerber, S.A.; Croy, B.A. Emerging themes in uterine natural killer cell heterogeneity and function. Am. J. Reprod. Immunol. 2012, 68, 282–289. [Google Scholar] [CrossRef]
  93. Sharma, S. Natural killer cells and regulatory T cells in early pregnancy loss. Int. J. Dev. Biol. 2014, 58, 219–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Arad, M.; Atzil, S.; Shakhar, K.; Adoni, A.; Ben-Eliyahu, S. Poly I-C induces early embryo loss in F344 rats: A potential role for NK cells. Am. J. Reprod. Immunol. 2005, 54, 49–53. [Google Scholar] [CrossRef] [PubMed]
  95. Reister, F.; Frank, H.G.; Kingdom, J.C.; Heyl, W.; Kaufmann, P.; Rath, W.; Huppertz, B. Macrophage-induced apoptosis limits endovascular trophoblast invasion in the uterine wall of preeclamptic women. Lab. Investig. 2001, 81, 1143–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Abrahams, V.M.; Kim, Y.M.; Straszewski, S.L.; Romero, R.; Mor, G. Macrophages and apoptotic cell clearance during pregnancy. Am. J. Reprod. Immunol. 2004, 51, 275–282. [Google Scholar] [CrossRef]
  97. Cotechini, T.; Komisarenko, M.; Sperou, A.; Macdonald-Goodfellow, S.; Adams, M.A.; Graham, C.H. Inflammation in rat pregnancy inhibits spiral artery remodeling leading to fetal growth restriction and features of preeclampsia. J. Exp. Med. 2014, 211, 165–179. [Google Scholar] [CrossRef]
  98. Phillips, P.; Brown, M.B.; Progulske-Fox, A.; Wu, X.J.; Reyes, L. Porphyromonas gingivalis strain-dependent inhibition of uterine spiral artery remodeling in the pregnant rat. Biol. Reprod. 2018, 99, 1045–1056. [Google Scholar] [CrossRef] [Green Version]
  99. Renaud, S.J.; Cotechini, T.; Quirt, J.S.; Macdonald-Goodfellow, S.K.; Othman, M.; Graham, C.H. Spontaneous pregnancy loss mediated by abnormal maternal inflammation in rats is linked to deficient uteroplacental perfusion. J. Immunol. 2011, 186, 1799–1808. [Google Scholar] [CrossRef] [Green Version]
  100. Falcón, B.J.; Cotechini, T.; Macdonald-Goodfellow, S.K.; Othman, M.; Graham, C.H. Abnormal inflammation leads to maternal coagulopathies associated with placental haemostatic alterations in a rat model of foetal loss. Thromb. Haemost. 2012, 107, 438–447. [Google Scholar] [CrossRef] [Green Version]
  101. Cotechini, T.; Hopman, W.J.; Graham, C.H. Inflammation-induced fetal growth restriction in rats is associated with altered placental morphometrics. Placenta 2014, 35, 575–581. [Google Scholar] [CrossRef]
  102. Gong, P.; Liu, M.; Hong, G.; Li, Y.; Xue, P.; Zheng, M.; Wu, M.; Shen, L.; Yang, M.; Diao, Z.; et al. Curcumin improves LPS-induced preeclampsia-like phenotype in rat by inhibiting the TLR4 signaling pathway. Placenta 2016, 41, 45–52. [Google Scholar] [CrossRef]
  103. Robb, K.P.; Cotechini, T.; Allaire, C.; Sperou, A.; Graham, C.H. Inflammation-induced fetal growth restriction in rats is associated with increased placental HIF-1α accumulation. PLoS ONE 2017, 12, e0175805. [Google Scholar] [CrossRef] [PubMed]
  104. Xue, P.; Zheng, M.; Gong, P.; Lin, C.; Zhou, J.; Li, Y.; Shen, L.; Diao, Z.; Yan, G.; Sun, H.; et al. Single administration of ultra-low-dose lipopolysaccharide in rat early pregnancy induces TLR4 activation in the placenta contributing to preeclampsia. PLoS ONE 2015, 10, e0124001. [Google Scholar] [CrossRef] [PubMed]
  105. Fan, M.; Li, X.; Gao, X.; Dong, L.; Xin, G.; Chen, L.; Qiu, J.; Xu, Y. LPS induces preeclampsia-like phenotype in rats and HTR8/SVneo cells dysfunction through TLR4/p38 MAPK pathway. Front. Physiol. 2019, 10, 1030. [Google Scholar] [CrossRef] [Green Version]
  106. Li, Z.H.; Wang, L.L.; Liu, H.; Muyayalo, K.P.; Huang, X.B.; Mor, G.; Liao, A.H. Galectin-9 alleviates LPS-induced preeclampsia-like impairment in rats via switching decidual macrophage polarization to M2 subtype. Front. Immunol. 2019, 9, 3142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Kozai, K.; Iqbal, K.; Moreno-Irusta, A.; Scott, R.L.; Simon, M.E.; Dhakal, P.; Fields, P.E.; Soares, M.J. Protective role of IL33 signaling in negative pregnancy outcomes associated with lipopolysaccharide exposure. FASEB J. 2021, 35, e21272. [Google Scholar] [CrossRef]
  108. Spaans, F.; Melgert, B.N.; Chiang, C.; Borghuis, T.; Klok, P.A.; de Vos, P.; van Goor, H.; Bakker, W.W.; Faas, M.M. Extracellular ATP decreases trophoblast invasion, spiral artery remodeling and immune cells in the mesometrial triangle in pregnant rats. Placenta 2014, 35, 587–595. [Google Scholar] [CrossRef] [PubMed]
  109. Waddell, B.J.; Hisheh, S.; Dharmarajan, A.M.; Burton, P.J. Apoptosis in rat placenta is zone dependent and stimulated by glucocorticoids. Biol. Reprod. 2000, 63, 1913–1917. [Google Scholar] [CrossRef]
  110. Ain, R.; Canham, L.N.; Soares, M.J. Dexamethasone-induced intrauterine growth restriction impacts the placental prolactin family, insulin-like growth factor-II and the Akt signaling pathway. J. Endocrinol. 2005, 185, 253–263. [Google Scholar] [CrossRef]
  111. Yokoi, R.; Suda, S.; Kobayashi, K.; Kuroda, J.; Kusama, H.; Kagami, H.; Ono, T. Differential susceptibility of rat embryos to methyl methanesulfonate during the pregastrulation period. J. Toxicol. Sci. 2007, 32, 495–503. [Google Scholar] [CrossRef] [Green Version]
  112. Furukawa, S.; Usuda, K.; Abe, M.; Hayashi, S.; Ogawa, I. Effect of 6-mercaptopurine on rat placenta. J. Vet. Med. Sci. 2008, 70, 551–556. [Google Scholar] [CrossRef] [Green Version]
  113. Furukawa, S.; Hayashi, S.; Usuda, K.; Abe, M.; Ogawa, I. The relationship between fetal growth restriction and small placenta in 6-mercaptopurine exposed rat. Exp. Toxicol. Pathol. 2011, 63, 89–95. [Google Scholar] [CrossRef] [PubMed]
  114. Taki, K.; Fukushima, T.; Ise, R.; Horii, I.; Yoshida, T. 6-Mercaptopurine-induced histopathological changes and xanthine oxidase expression in rat placenta. J. Toxicol. Sci. 2012, 37, 607–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Furukawa, S.; Hayashi, S.; Usuda, K.; Abe, M.; Hagio, S.; Ogawa, I. Effect of cisplatin on rat placenta development. Exp. Toxicol. Pathol. 2013, 65, 211–217. [Google Scholar] [CrossRef] [PubMed]
  116. Sun, J.; Sugiyama, A.; Inoue, S.; Takeuchi, T.; Takeuchi, T.; Furukawa, S. Effect of methotrexate on rat placenta development. Exp. Toxicol. Pathol. 2013, 65, 995–1002. [Google Scholar] [CrossRef]
  117. Furukawa, S.; Hayashi, S.; Abe, M.; Hagio, S.; Irie, K.; Kuroda, Y.; Ogawa, I.; Sugiyama, A. Effect of chlorpromazine on rat placenta development. Exp. Toxicol. Pathol. 2014, 66, 41–47. [Google Scholar] [CrossRef]
  118. Lee, C.K.; Lee, C.U.; Kim, J.H.; Son, B.C.; Kim, D.H.; Lee, C.H.; Kim, H.D.; Kim, J.W.; Yoon, Y.D.; Kang, S.G.; et al. Effects of Aroclor 1254 on the expression of rat placental PRL-family genes. Mol. Cells. 2003, 15, 114–121. [Google Scholar]
  119. Nishimura, K.; Nakano, N.; Chowdhury, V.S.; Kaneto, M.; Torii, M.; Hattori, M.A.; Yamauchi, N.; Kawai, M. Effect of PPARβ/δ agonist on the placentation and embryo-fetal development in rats. Birth Defects Res. B Dev. Reprod. Toxicol. 2013, 98, 164–169. [Google Scholar] [CrossRef]
  120. Holloway, A.C.; Salomon, X.A.; Soares, M.J.; Garnier, V.; Raha, S.; Sergent, F.; Nicholson, C.J.; Feige, J.J.; Benharouga, M.; Alfaidy, N. Characterization of the adverse effects of nicotine on placental development: In vivo and in vitro studies. Am. J. Physiol. Endocrinol. Metab. 2014, 306, E443–E456. [Google Scholar] [CrossRef] [Green Version]
  121. Silva, J.F.; Ocarino, N.M.; Serakides, R. Maternal thyroid dysfunction affects placental profile of inflammatory mediators and the intrauterine trophoblast migration kinetics. Reproduction 2014, 147, 803–816. [Google Scholar] [CrossRef]
  122. Chakraborty, S.; Islam, S.; Saha, S.; Ain, R. Dexamethasone-induced intra-uterine growth restriction impacts nostrin and its downstream effector genes in the rat mesometrial uterus. Sci. Rep. 2018, 8, 8342. [Google Scholar] [CrossRef] [Green Version]
  123. Furukawa, S.; Tsuji, N.; Hayashi, S.; Kuroda, Y.; Kimura, M.; Hayakawa, C.; Takeuchi, K.; Sugiyama, A. The effects of β-naphthoflavone on rat placental development. J. Toxicol. Pathol. 2019, 32, 275–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Furukawa, S.; Hayashi, S.; Usuda, K.; Abe, M.; Ogawa, I. The impairment of metrial gland development in tamoxifen exposed rats. Exp. Toxicol. Pathol. 2012, 64, 121–126. [Google Scholar] [CrossRef] [PubMed]
  125. Iqbal, K.; Pierce, S.H.; Kozai, K.; Dhakal, P.; Scott, R.L.; Roby, K.F.; Vyhlidal, C.A.; Soares, M.J. Evaluation of placentation and the role of the aryl hydrocarbon receptor pathway in a rat model of dioxin exposure. Environ. Health Perspect. 2021, 129, 117001. [Google Scholar] [CrossRef] [PubMed]
  126. Tai, M.; Piskorski, A.; Kao, J.C.; Hess, L.A.; de la Monte, S.; Gündoğan, F. Placental Morphology in Fetal Alcohol Spectrum Disorders. Alcohol Alcohol. 2017, 52, 138–144. [Google Scholar] [CrossRef]
  127. Gundogan, R.; Gilligan, J.; Ooi, J.-H.; Sung, J.; Qi, W.; Naram, R.; de la Monte, S.M. Dual mechanisms of ethanol-impaired placentation: Experimental model. Clin. Exper. Pathol. 2013, 3, 2. [Google Scholar] [CrossRef]
  128. Gårdebjer, E.M.; Cuffe, J.S.; Pantaleon, M.; Wlodek, M.E.; Moritz, K.M. Periconceptional alcohol consumption causes fetal growth restriction and increases glycogen accumulation in the late gestation rat placenta. Placenta 2014, 35, 50–57. [Google Scholar] [CrossRef]
  129. Gundogan, F.; Elwood, G.; Longato, L.; Tong, M.; Feijoo, A.; Carlson, R.I.; Wands, J.R.; de la Monte, S.M. Impaired placentation in fetal alcohol syndrome. Placenta 2008, 29, 148–157. [Google Scholar] [CrossRef] [Green Version]
  130. Gundogan, F.; Gilligan, J.; Qi, W.; Chen, E.; Naram, R.; de la Monte, S.M. Dose effect of gestational ethanol exposure on placentation and fetal growth. Placenta 2015, 36, 523–530. [Google Scholar] [CrossRef] [Green Version]
  131. Akay, M.T.; Kockaya, E.A. The effects of alcohol on rat placenta. Cell Biochem. Funct. 2005, 23, 435–445. [Google Scholar]
  132. Furukawa, S.; Hayashi, S.; Usuda, K.; Abe, M.; Ogawa, I. Histopathological effect of ketoconazole on rat placenta. J. Vet. Med. Sci. 2008, 70, 1179–1184. [Google Scholar] [CrossRef] [Green Version]
  133. Estrada-Flores, E.; Yáñez Mendoza, C. Histology of rat placentas treated with methylhydrazine Ro 4-6467. Bol. Estud. Med. Biol. 1990, 38, 59–64. [Google Scholar] [PubMed]
  134. Ahbab, M.A.; Güven, C.; Koçkaya, E.A.; Barlas, N. Comparative developmental toxicity evaluation of di- n-hexyl phthalate and dicyclohexyl phthalate in rats. Toxicol. Ind. Health 2017, 33, 696–716. [Google Scholar] [CrossRef] [PubMed]
  135. Jawerbaum, A.; White, V. Animal models in diabetes and pregnancy. Endocr. Rev. 2010, 31, 680–701. [Google Scholar] [CrossRef] [Green Version]
  136. Gewolb, I.H.; Merdian, W.; Warshaw, J.B.; Enders, A.C. Fine structural abnormalities of the placenta in diabetic rats. Diabetes 1986, 35, 1254–1261. [Google Scholar] [CrossRef]
  137. Ne’eman, Z.; Barash, V.; Rosenmann, E.; Shafrir, E. Localization of glycogen in the placenta of diabetic rats: A light and electron microscopic study. Placenta 1987, 8, 201–208. [Google Scholar] [CrossRef]
  138. Zorn, T.M.; Zúñiga, M.; Madrid, E.; Tostes, R.; Fortes, Z.; Giachini, F.; San Martín, S. Maternal diabetes affects cell proliferation in developing rat placenta. Histol. Histopathol. 2011, 26, 1049–1056. [Google Scholar] [PubMed]
  139. Farias, P.S.; dos SSouza, K.; Fioretto, E.T.; dos Santos, M.R.; Aires, M.B. Maternal diabetes affects rat placental morphology and pregnancy. Endocrine 2014, 45, 497–501. [Google Scholar] [CrossRef] [PubMed]
  140. Nteeba, J.; Varberg, K.M.; Scott, R.L.; Simon, M.E.; Iqbal, K.; Soares, M.J. Poorly controlled diabetes mellitus alters placental structure, efficiency, and plasticity. BMJ Open Diabetes Res. Care 2020, 8, e001243. [Google Scholar] [CrossRef]
  141. Phillips, J.K.; Vance, A.M.; Raj, R.S.; Mandalà, M.; Linder, E.A.; Gokina, N.I. Impact of experimental diabetes on the maternal uterine vascular remodeling during rat pregnancy. Reprod. Sci. 2012, 19, 322–331. [Google Scholar] [CrossRef] [Green Version]
  142. Fisher, S.J. Why is placentation abnormal in preeclampsia? Am. J. Obstet. Gynecol. 2015, 213, S115–S122. [Google Scholar] [CrossRef] [Green Version]
  143. Bakrania, B.A.; George, E.M.; Granger, J.P. Animal models of preeclampsia: Investigating pathophysiology and therapeutic targets. Am. J. Obstet. Gynecol. 2020, 226, S973–S987. [Google Scholar] [CrossRef] [PubMed]
  144. Rana, S.; Lemoine, E.; Granger, J.P.; Karumanchi, S.A. Preeclampsia: Pathophysiology, challenges, and perspectives. Circ. Res. 2019, 124, 1094–1112. [Google Scholar] [CrossRef] [PubMed]
  145. Croskerry, P.G.; Smith, G.K.; Hall, S.; Shepard, B.J. Fetal and placental growth in the rat following differential reduction of litter size. Biol. Neonate 1978, 33, 31–38. [Google Scholar] [CrossRef]
  146. vom Saal, F.S. Variation in phenotype due to random intrauterine positioning of male and female fetuses in rodents. J. Reprod. Fertil. 1981, 62, 633–650. [Google Scholar] [CrossRef] [Green Version]
  147. McLaurin, K.A.; Mactutus, C.F. Polytocus focus: Uterine position effect is dependent upon horn size. Int. J. Dev. Neurosci. 2015, 40, 85–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Nüsken, K.D.; Warnecke, C.; Hilgers, K.F.; Schneider, H. Intrauterine growth after uterine artery ligation in rats: Dependence on the fetal position in the uterine horn and need for prenatal marking of the animals. J. Hypertens. 2007, 25, 247–248. [Google Scholar] [CrossRef]
  149. Bohlender, J.; Ganten, D.; Luft, F.C. Rats transgenic for human renin and human angiotensinogen as a model for gestational hypertension. J. Am. Soc. Nephrol. 2000, 11, 2056–2061. [Google Scholar] [CrossRef]
  150. Dechend, R.; Gratze, P.; Wallukat, G.; Shagdarsuren, E.; Plehm, R.; Bräsen, J.H.; Fiebeler, A.; Schneider, W.; Caluwaerts, S.; Vercruysse, L.; et al. Agonistic autoantibodies to the AT1 receptor in a transgenic rat model of preeclampsia. Hypertension 2005, 45, 742–746. [Google Scholar] [CrossRef] [Green Version]
  151. Verlohren, S.; Niehoff, M.; Hering, L.; Geusens, N.; Herse, F.; Tintu, A.N.; Plagemann, A.; LeNoble, F.; Pijnenborg, R.; Muller, D.N.; et al. Uterine vascular function in a transgenic preeclampsia rat model. Hypertension 2008, 51, 547–553. [Google Scholar] [CrossRef] [Green Version]
  152. Geusens, N.; Verlohren, S.; Luyten, C.; Taube, M.; Hering, L.; Vercruysse, L.; Hanssens, M.; Dudenhausen, J.W.; Dechend, R.; Pijnenborg, R. Endovascular trophoblast invasion, spiral artery remodelling and uteroplacental haemodynamics in a transgenic rat model of pre-eclampsia. Placenta 2008, 29, 614–623. [Google Scholar] [CrossRef]
  153. Geusens, N.; Hering, L.; Verlohren, S.; Luyten, C.; Drijkoningen, K.; Taube, M.; Vercruysse, L.; Hanssens, M.; Dechend, R.; Pijnenborg, R. Changes in endovascular trophoblast invasion and spiral artery remodelling at term in a transgenic preeclamptic rat model. Placenta 2010, 31, 320–326. [Google Scholar] [CrossRef] [PubMed]
  154. Hering, L.; Herse, F.; Geusens, N.; Verlohren, S.; Wenzel, K.; Staff, A.C.; Brosnihan, K.B.; Huppertz, B.; Luft, F.C.; Muller, D.N.; et al. Effects of circulating and local uteroplacental angiotensin II in rat pregnancy. Hypertension 2010, 56, 311–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Golic, M.; Haase, N.; Herse, F.; Wehner, A.; Vercruysse, L.; Pijnenborg, R.; Balogh, A.; Saether, P.C.; Dissen, E.; Luft, F.C.; et al. Natural killer cell reduction and uteroplacental vasculopathy. Hypertension 2016, 68, 964–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Okamoto, K.; Aoki, K. Development of a strain of spontaneously hypertensive rats. Jpn. Circ. J. 1963, 27, 282–293. [Google Scholar] [CrossRef] [PubMed]
  157. Barrientos, G.; Pussetto, M.; Rose, M.; Staff, A.C.; Blois, S.M.; Toblli, J.E. Defective trophoblast invasion underlies fetal growth restriction and preeclampsia-like symptoms in the stroke-prone spontaneously hypertensive rat. Mol. Hum. Reprod. 2017, 23, 509–519. [Google Scholar] [CrossRef]
  158. Li, J.; LaMarca, B.; Reckelhoff, J.F. A model of preeclampsia in rats: The reduced uterine perfusion pressure (RUPP) model. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H1–H8. [Google Scholar] [CrossRef]
  159. Coats, L.E.; Bamrick-Fernandez, D.R.; Ariatti, A.M.; Bakrania, B.A.; Rawls, A.Z.; Ojeda, N.B.; Alexander, B.T. Stimulation of soluble guanylate cyclase diminishes intrauterine growth restriction in a rat model of placental ischemia. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2021, 320, R149–R161. [Google Scholar] [CrossRef]
  160. Podjarny, E.; Bernheim, J.; Katz, B.; Green, J.; Mekler, J.; Bursztyn, M. Chronic exogenous hyperinsulinaemia in pregnancy: A rat model of pregnancy-induced hypertension. J. Am. Soc. Nephrol. 1998, 9, 9–13. [Google Scholar] [CrossRef]
  161. Skarzinski, G.; Khamaisi, M.; Bursztyn, M.; Mekler, J.; Lan, D.; Evdokimov, P.; Ariel, I. Intrauterine growth restriction and shallower implantation site in rats with maternal hyperinsulinemia are associated with altered NOS expression. Placenta 2009, 30, 898–906. [Google Scholar] [CrossRef]
  162. Kreiser, D.; Nguyen, X.; Wong, R.; Seidman, D.; Stevenson, D.; Quan, S.; Abraham, N.; Dennery, P.A. Heme oxygenase-1 modulates fetal growth in the rat. Lab. Investig. 2002, 82, 687–692. [Google Scholar] [CrossRef] [Green Version]
  163. Oltra, L.; Reverte, V.; Garcés, B.; Li Volti, G.; Moreno, J.M.; Salazar, F.J.; Llinás, M.T. Trophoblast-induced spiral artery remodelling and uteroplacental haemodynamics in pregnant rats with increased blood pressure induced by heme oxygenase inhibition. Placenta 2020, 89, 91–98. [Google Scholar] [CrossRef] [PubMed]
  164. Hirschi, K.M.; Tsai, K.Y.; Davis, T.; Clark, J.C.; Knowlton, M.N.; Bikman, B.T.; Reynolds, P.R.; Arroyo, J.A. Growth arrest-specific protein-6/AXL signaling induces preeclampsia in rats. Biol. Reprod. 2020, 102, 199–210. [Google Scholar] [CrossRef] [PubMed]
  165. Belkacemi, L.; Desai, M.; Nelson, D.M.; Ross, M.G. Altered mitochondrial apoptotic pathway in placentas from undernourished rat gestations. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 301, R1599–R1615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Rebelato, H.J.; Esquisatto, M.A.; de Sousa Righi, E.F.; Catisti, R. Gestational protein restriction alters cell proliferation in rat placenta. J. Mol. Histol. 2016, 47, 203–211. [Google Scholar] [CrossRef]
  167. Padmanabhan, R.; Al-Menhali, N.M.; Ahmed, I.; Kataya, H.H.; Ayoub, M.A. Histological, histochemical and electron microscopic changes of the placenta induced by maternal exposure to hyperthermia in the rat. Int. J. Hyperth. 2005, 21, 29–44. [Google Scholar] [CrossRef]
  168. Roberts, H.; Woodman, A.G.; Baines, K.J.; Jeyarajah, M.J.; Bourque, S.L.; Renaud, S.J. Maternal iron deficiency alters trophoblast differentiation and placental development in rat pregnancy. Endocrinology 2021, 162, bqab215. [Google Scholar] [CrossRef]
  169. Mark, P.J.; Sisala, C.; Connor, K.; Patel, R.; Lewis, J.L.; Vickers, M.H.; Waddell, B.J.; Sloboda, D.M. A maternal high-fat diet in rat pregnancy reduces growth of the fetus and the placental junctional zone, but not placental labyrinth zone growth. J. Dev. Origins Health Dis. 2011, 2, 63–70. [Google Scholar] [CrossRef]
  170. Hayes, E.K.; Tessier, D.R.; Percival, M.E.; Holloway, A.C.; Petrik, J.J.; Gruslin, A.; Raha, S. Trophoblast invasion and blood vessel remodeling are altered in a rat model of lifelong maternal obesity. Reprod. Sci. 2014, 21, 648–657. [Google Scholar] [CrossRef] [Green Version]
  171. Faria, T.N.; Soares, M.J. 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. [Google Scholar] [CrossRef]
  172. 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, 50–63.e6. [Google Scholar] [CrossRef] [Green Version]
  173. Arroyo, J.A.; Konno, T.; Khalili, D.C.; Soares, M.J. A simple in vivo approach to investigate invasive trophoblast cells. Int. J. Dev. Biol. 2005, 49, 977–980. [Google Scholar] [CrossRef] [PubMed]
  174. Lee, D.S.; Rumi, M.A.; Konno, T.; Soares, M.J. In vivo genetic manipulation of the rat trophoblast cell lineage using lentiviral vector delivery. Genesis 2009, 47, 433–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Chakraborty, D.; Muto, M.; Soares, M.J. Ex vivo trophoblast-specific genetic manipulation using lentiviral delivery. Bio. Protoc. 2017, 7, e2652. [Google Scholar] [CrossRef] [Green Version]
  176. Varberg, K.M.; Iqbal, K.; Muto, M.; Simon, M.E.; Scott, R.L.; Kozai, K.; Choudhury, R.H.; Aplin, J.D.; Biswell, R.; Gibson, M.; et al. ASCL2 reciprocally controls key trophoblast lineage decisions during hemochorial placenta development. Proc. Natl. Acad. Sci. USA 2021, 118, e2016517118. [Google Scholar] [CrossRef] [PubMed]
  177. Kent, L.N.; Konno, T.; Soares, M.J. Phosphatidylinositol 3 kinase modulation of trophoblast cell differentiation. BMC Dev. Biol. 2010, 10, 97. [Google Scholar] [CrossRef] [Green Version]
  178. Kent, L.N.; Rumi, M.A.; Kubota, K.; Lee, D.S.; Soares, M.J. FOSL1 is integral to establishing the maternal-fetal interface. Mol. Cell Biol. 2011, 31, 4801–4813. [Google Scholar] [CrossRef] [Green Version]
  179. Kubota, K.; Kent, L.N.; Rumi, M.A.; Roby, K.F.; Soares, M.J. Dynamic regulation of AP-1 transcriptional complexes directs trophoblast differentiation. Mol. Cell Biol. 2015, 35, 3163–3177. [Google Scholar] [CrossRef] [Green Version]
  180. Renaud, S.J.; Kubota, K.; Rumi, M.A.; Soares, M.J. The FOS transcription factor family differentially controls trophoblast migration and invasion. J. Biol. Chem. 2014, 289, 5025–5039. [Google Scholar] [CrossRef] [Green Version]
  181. Amoroso, E.C. The evolution of viviparity. Proc. R. Soc. Med. 1968, 61, 1188–1200. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representations of the rat hemochorial placentation site during gestation. (A) Relationship of natural killer (NK) cells and invasive trophoblast cells in transformation of the uterus, including the uterine vasculature. (B) Structural compartments and cellular constituents of the mature rat placentation site.
Figure 1. Schematic representations of the rat hemochorial placentation site during gestation. (A) Relationship of natural killer (NK) cells and invasive trophoblast cells in transformation of the uterus, including the uterine vasculature. (B) Structural compartments and cellular constituents of the mature rat placentation site.
Ijms 23 02947 g001
Figure 2. Schematic representations of hemochorial placentation sites of the human and rat highlighting similarities in structural organization. The extravillous trophoblast column and junctional zone are homologous structures of the human and rat, respectively. These structures are the sources of trophoblast cells that invade deep into the uterus. The villous and labyrinth compartments are homologous structures of the human and rat, respectively, and represent the site of maternal-fetal exchange. Histological organization of the villous and labyrinth compartments are defined as hemomonochorial and hemotrichorial, respectively.
Figure 2. Schematic representations of hemochorial placentation sites of the human and rat highlighting similarities in structural organization. The extravillous trophoblast column and junctional zone are homologous structures of the human and rat, respectively. These structures are the sources of trophoblast cells that invade deep into the uterus. The villous and labyrinth compartments are homologous structures of the human and rat, respectively, and represent the site of maternal-fetal exchange. Histological organization of the villous and labyrinth compartments are defined as hemomonochorial and hemotrichorial, respectively.
Ijms 23 02947 g002
Figure 3. Infiltration of invasive trophoblast cells into the uterine-placental interface during the second half of gestation. Invasive trophoblast cells were identified in placentation sites of wild-type female rats mated with males possessing a chicken β-actin promoter-enhanced green fluorescence protein transgene. Green fluorescence reflects trophoblast cells within the placenta and invasive trophoblast cells which have migrated into the uterine-placental interface. Note the expansion of invasive trophoblast cells into the uterine-placental interface as gestation progresses. Abbreviations: UPI, uterine-placental interface; Dec, decidua; JZ, junctional zone; LZ, labyrinth zone. (Adapted from Reference [46]).
Figure 3. Infiltration of invasive trophoblast cells into the uterine-placental interface during the second half of gestation. Invasive trophoblast cells were identified in placentation sites of wild-type female rats mated with males possessing a chicken β-actin promoter-enhanced green fluorescence protein transgene. Green fluorescence reflects trophoblast cells within the placenta and invasive trophoblast cells which have migrated into the uterine-placental interface. Note the expansion of invasive trophoblast cells into the uterine-placental interface as gestation progresses. Abbreviations: UPI, uterine-placental interface; Dec, decidua; JZ, junctional zone; LZ, labyrinth zone. (Adapted from Reference [46]).
Ijms 23 02947 g003
Table 1. Experimental manipulations impacting the rat uterine-placental interface.
Table 1. Experimental manipulations impacting the rat uterine-placental interface.
Experimental Maternal ManipulationPhenotype of the Uterine-Placental InterfaceReferences
Oxygen
Moderate hypoxia during placental morphogenesisExpansion of the junctional zone; enhanced endovascular invasive trophoblast restructuring of uterine spiral arteries[46,63,64]
Severe/chronic hypoxiaImpairments in junctional zone development and trophoblast-guided spiral artery remodeling[70,71,72,73,74]
Immune cells
NK cellsRemodel uterine arterial vessels; restrain intrauterine endovascular trophoblast cell invasion [47,90]
MacrophagesUterine angiogenesis, tissue repair, artery remodeling, immune tolerance, postpartum trophoblast removal [78,83,85,86,87,88,89]
Drug, toxicant, and miscellaneous exposure
Anti-cancer drugs, dexa-methasone, AHR agonists, GW501516, nicotine Hypoplasia of the junctional zone, abnormalities in glycogen trophoblast cell development, and/or decreased intrauterine interstitial trophoblast cell invasion [109,110,111,112,113,114,115,116,117,118,119,120,121,122,123]
TamoxifenDisrupts the uterine-placental interface; decreases NK cell numbers; defective uterine spiral artery transformation [124]
DoxycyclineImpairs endovascular trophoblast cell invasion and uterine spiral artery remodeling and placental perfusion[82]
DioxinAccelerates endovascular trophoblast cell invasion[125]
Chronic ethanolDecreased glycogen trophoblast cell numbers; shallow trophoblast invasion; failed uterine spiral artery remodeling[127,129,130,131]
Ketoconazole, methylhydrazineExpansion of junctional zone and glycogen trophoblast cell clusters[132,133]
Disease states
Diabetes: assorted hyperglycemia modelsJunctional zone and glycogen trophoblast cell expansion; impaired trophoblast invasion and spiral artery remodeling; retention/expansion of NK cells and macrophages [136,137,138,139,140,141]
Hypertension/preeclampsia:
Angiotensinogen-renin transgenic model
Enhanced endovascular trophoblast cell invasion and uterine spiral artery remodeling[152,153,154]
Hypertension/preeclampsia:
Stroke prone spontaneous hypertensive rat (SHSRP)
Impaired junctional zone and glycogen trophoblast cell development; shallow trophoblast cell invasion; failed uterine spiral artery remodeling[157]
Hypertension/preeclampsia:
Reduced uterine perfusion pressure (RUPP) model
Expanded junctional zone and impairments in intrauterine trophoblast cell invasion and uterine spiral artery remodeling[72,158,159]
Hypertension/preeclampsia:
Maternal hyperinsulinemia
Enhanced endovascular trophoblast cell invasion[161]
Hypertension/preeclampsia:
Hemoxygenase inhibition
Diminished endovascular trophoblast cell-guided uterine spiral artery remodeling[163]
Hypertension/preeclampsia:
GAS6 treatment
Decreased interstitial trophoblast cell invasion[164]
Malnutrition/protein restriction/hyperthermiaNegative impact on junctional zone development[165,166,167]
Iron deficiencyExpanded junctional zone[168]
High fat dietDiminished junctional zone; early increase in intrauterine trophoblast invasion; ate gestation decline in interstitial trophoblast cell invasion[169,170]
Table 2. Regulatory genes affecting the rat uterine-placental interface.
Table 2. Regulatory genes affecting the rat uterine-placental interface.
GenesMutationPhenotypeReferences
Fosl1Trophoblast-LOF aInhibition of endovascular trophoblast cell invasion[178]
Kdm3aTrophoblast-LOFDisruption of hypoxia-activated endovascular trophoblast cell invasion[64]
Mmp12Global-LOFDisruption of hypoxia-activated endovascular trophoblast cell invasion[64]
Ascl2Global-LOFJunctional zone dysgenesis and failed intrauterine trophoblast cell invasion[176]
AhrGlobal-LOFMaternal AHR regulates TCDD-induced endovascular trophoblast cell invasion[125]
TfpiTrophoblast-LOFInhibition of endovascular and interstitial trophoblast cell invasion[48]
a LOF, loss of function.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shukla, V.; Soares, M.J. Modeling Trophoblast Cell-Guided Uterine Spiral Artery Transformation in the Rat. Int. J. Mol. Sci. 2022, 23, 2947. https://doi.org/10.3390/ijms23062947

AMA Style

Shukla V, Soares MJ. Modeling Trophoblast Cell-Guided Uterine Spiral Artery Transformation in the Rat. International Journal of Molecular Sciences. 2022; 23(6):2947. https://doi.org/10.3390/ijms23062947

Chicago/Turabian Style

Shukla, Vinay, and Michael J. Soares. 2022. "Modeling Trophoblast Cell-Guided Uterine Spiral Artery Transformation in the Rat" International Journal of Molecular Sciences 23, no. 6: 2947. https://doi.org/10.3390/ijms23062947

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop