Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2889 2022-05-10 14:34:47 |
2 format -22 word(s) 2867 2022-05-11 03:43:44 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Shukla, V.; Soares, M. Trophoblast Cell Invasion and Uterine Spiral Artery Remodeling. Encyclopedia. Available online: https://encyclopedia.pub/entry/22772 (accessed on 15 May 2024).
Shukla V, Soares M. Trophoblast Cell Invasion and Uterine Spiral Artery Remodeling. Encyclopedia. Available at: https://encyclopedia.pub/entry/22772. Accessed May 15, 2024.
Shukla, Vinay, Michael Soares. "Trophoblast Cell Invasion and Uterine Spiral Artery Remodeling" Encyclopedia, https://encyclopedia.pub/entry/22772 (accessed May 15, 2024).
Shukla, V., & Soares, M. (2022, May 10). Trophoblast Cell Invasion and Uterine Spiral Artery Remodeling. In Encyclopedia. https://encyclopedia.pub/entry/22772
Shukla, Vinay and Michael Soares. "Trophoblast Cell Invasion and Uterine Spiral Artery Remodeling." Encyclopedia. Web. 10 May, 2022.
Trophoblast Cell Invasion and Uterine Spiral Artery Remodeling
Edit

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. 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.

Trophoblast Cell Uterine Spiral Artery Remodeling Oxygen Immune Cells

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].

2. Oxygen Tension

Oxygen is an essential cellular nutrient and a critical regulator of hemochorial placentation [14][15]. Trophoblast cell development occurs in a low oxygen environment [14] and is under the control of hypoxia signaling pathways [15][16][17]. 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 [18][19] and triggering enhanced development of endovascular invasive trophoblast cells, and their invasion, colonization, and restructuring of uterine spiral arteries [18]. 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) [20]. In response to low oxygen tension HIF activates the transcription of a cohort of genes, including KDM3A [20][21]. The trophoblast cell epigenetic landscape is redirected through the demethylating actions of KDM3A on histone 3 lysine 9 (H3K9), leading to further gene activation [20]. Among the genes activated in endovascular invasive trophoblast cells by this pathway is MMP12 [20][22]. Arterial elastin is degraded by MMP12 resulting in restructuring uterine spiral arteries [20][23][24]. The HIF-KDM3A-MMP12 pathway was elucidated using rat trophoblast stem (TS) cells and in vivo genetically manipulated rat models [20][25] and conservation of elements of the pathway determined using human placental tissue specimens and human trophoblast cells [20][22][23][24][26].
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 [27][28][29][30][31]. These adverse effects may be mediated via enothelin-1 signaling [27][31] and disruptions in matrix metallopeptidase expression [29]. Adverse consequences of poor oxygenation also emerge from failed placentation and compromised ability of the placenta to adapt to hypoxia [14][32][33]. Enhancing oxygen delivery can rescue some of the unfavorable effects of placental hypoxia [30].

3. 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 [34][35][36][37]. The uterine-placental interface exhibits dynamic changes in NK cell abundance, which shows a reciprocal relationship to the presence of invasive trophoblast cells [35][38][39]. 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 [38][39]. 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 [40][41][42][43]. An exception is NK cell expansion in failed conceptus sites [44]. Macrophages are situated throughout the uterine-placental interface, including in locations proximal to uterine spiral arteries [45][46] and exhibit a pronounced increase in number at parturition [47][48][49].
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 [21][50]. Three key observations were made from pregnancies deficient in NK cells [21][50]: (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 [21][51]. 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 [21][38][50][52]. The nature of NK cell restraint on endovascular trophoblast cell invasion may be linked to oxygen delivery [21]. 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 [21][50]. 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 [53][54][55].
Macrophages possess several roles at the uterine-placental interface [35][36][37]. They contribute to the regulation of angiogenesis, post-implantation uterine tissue repair, uterine spiral artery remodeling, and immune tolerance [36][37]. Most interestingly, they expand in number at term [47][48][49] and have been hypothesized to contribute to post-partum reconstruction of the uterus, including removal of invasive trophoblast cells [36][37][56][57][58]. Macrophages are also linked to pregnancy-dependent uterine-placental interface responses to inflammation and infection [59][60]. 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 [59][61][62][63][64][65]. TLR4 activation and tumor necrosis factor alpha are viewed as drivers of the deleterious actions of LPS on the uterine-placental interface [59][66][67], whereas galectins and interleukin 33 can protect against the unfavorable consequences of LPS exposure [68][69]. 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 [70]. ATP-induced macrophage activation results in restricted interstitial trophoblast cell invasion and impaired uterine spiral artery remodeling [70]. 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 [60].

4. 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 [71]. 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 [72][73][74][75][76][77][78][79][80][81][82][83][84][85][86]. 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 [87]. In vivo treatment with doxycycline, a tetracycline antibiotic, impairs endovascular trophoblast cell invasion and uterine spiral artery remodeling and placental perfusion [42]. The mode of action of doxycycline may be through inhibition of matrix metallopeptidase activities [42]. 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 [88]. Interestingly, in the rat the actions of TCDD are not directly on trophoblast cells [88]. 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 [86]. 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 [88].
Maternal alcohol ingestion during pregnancy can lead to congenital anomalies, including placental and fetal growth restriction [89]. Timing and duration of ethanol exposure differentially affect placentation [90][91]. Chronic maternal ethanol intake during pregnancy in the rat adversely impacts placentation resulting in disruptions in the junctional zone [92][93][94], decreased prevalence of glycogen trophoblast cells, shallow intrauterine trophoblast cell invasion, and failure of uterine spiral artery remodeling [90][92][93]. 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 [92][93]. In contrast, transient maternal ethanol intake around the time of conception leads to an expansion of the junctional zone and glycogen trophoblast cells [91]. 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 [95][96]. Administration of phthalates, industrial plasticizers, to pregnant rats had contradictory actions [97]. 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. Disease States

An assortment of disease states affecting pregnancy and placentation can be modeled in the rat.

5.1. Diabetes

Diabetes during pregnancy has been simulated in the rat leading to maternal hyperglycemia with consequential effects on placental and fetal development [98]. Placentomegaly is a typical response to maternal hyperglycemia [99][100][101][102][103]. The junctional zone expands in size and glycogen trophoblast cell clusters become more abundant [99][100][101][102], which are characterized by prominent changes in the junctional zone transcriptome [103]. These events within the placentation site are connected to failures in intrauterine trophoblast cell invasion, especially the interstitial invasive trophoblast cell lineage [43][103], and impairments in uterine spiral artery remodeling [104]. Maternal hyperglycemia is also associated with a retention/expansion of NK cells and macrophages within the uterine-placental interface [43] and compromised plasticity of placentation to environmental stressors such as hypoxia [103]. Some of the consequences of maternal hyperglycemia may be mediated by the direct actions of elevated glucose on trophoblast cell development [103]. 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.

5.2. 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][105]. The literature is replete with descriptions of rat “models” of preeclampsia [106]. 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 [106][107]. Prevailing evidence indicates that failures in trophoblast cell-guided uterine spiral artery are the underlying cause of early onset preeclampsia in humans [10][105]. 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 [108][109][110][111]. 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. 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 [112][113][114]. Maternal hypertension arises from mating female rats expressing an angiotensinogen transgene with male rats expressing a human renin transgene [112]. In contrast to predictions based on preeclampsia in humans, this model of preeclampsia displayed enhanced endovascular trophoblast cell invasion and uterine spiral artery remodeling [115][116][117]. Differences in endovascular trophoblast cell invasion were not as pronounced by the end of gestation [116]. 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 [18]. 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 [117]. NK cells provide a protective role in this maternal hypertension model [118]. 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 [118].
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 [119]. Unlike the transgenic model described above, hypertension in the SHRSP rat is not restricted to pregnancy [120]. SHRSP pregnancies are characterized by small litter sizes and placental and fetal growth restriction [120]. 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 [120]. 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 [106][121]. 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 [121]. The RUPP procedure leads to an expanded junctional zone [122] and impairments in both intrauterine trophoblast invasion and uterine spiral artery remodeling [29].
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) [123][124][125][126][127]. 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 [124], whereas HO inhibition led to diminished endovascular trophoblast cell-guided uterine spiral artery remodeling [126]. In contrast, endovascular invasive trophoblast cells were not a target of exogenous GAS6 injections but instead, intrauterine interstitial trophoblast cell invasion was diminished [127]. The range of phenotypic responses to these experimental manipulations reflect the complexity of regulatory events controlling intrauterine trophoblast cell invasion and uterine vascular remodeling.

5.3. Malnutrition/Obesity/Hyperthermia

The rat uterine-placental interface has been examined following manipulations of diet and other pathophysiologic challenges. Again, the 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 [128][129][130]. In contrast, maternal iron deficiency resulted in an expansion of the size of the junctional zone [131]. 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 [132], an early increase in endovascular and interstitial trophoblast cell invasion and later decreased interstitial trophoblast cell invasion [133].

References

  1. Burton, G.J.; Fowden, A.L.; Thornburg, K.L. Placental origins of chronic disease. Physiol. Rev. 2016, 96, 1509–1565.
  2. Maltepe, E.; Fisher, S.J. Placenta: The forgotten organ. Annu. Rev. Cell. Dev. Biol. 2015, 31, 523–552.
  3. Roberts, R.M.; Green, J.A.; Schulz, L.C. The evolution of the placenta. Reproduction 2016, 152, R179–R189.
  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.
  5. Soares, M.J.; Varberg, K.M.; Iqbal, K. Hemochorial placentation: Development, function, and adaptations. Biol. Reprod. 2018, 99, 196–211.
  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.
  7. Pijnenborg, R.; Vercruysse, L.; Hanssens, M. The uterine spiral arteries in human pregnancy: Facts and controversies. Placenta 2006, 27, 939–958.
  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.
  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.
  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.
  11. Wooding, P.; Burton, G. Comparative Placentation; Springer: Heidelberg, Germany, 2008.
  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.
  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.
  14. Burton, G.J. Oxygen, the Janus gas; its effects on human placental development and function. J. Anat. 2009, 215, 27–35.
  15. Fryer, B.H.; Simon, M.C. Hypoxia, HIF and the placenta. Cell Cycle 2006, 5, 495–498.
  16. Soares, M.J.; Iqbal, K.; Kozai, K. Hypoxia and placental development. Birth Defects Res. 2017, 109, 1309–1329.
  17. 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.
  18. Rosario, G.X.; Konno, T.; Soares, M.J. Maternal hypoxia activates endovascular trophoblast cell invasion. Dev. Biol. 2008, 314, 362–375.
  19. 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.
  20. 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.
  21. 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.
  22. 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.
  23. 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.
  24. 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.
  25. 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.
  26. 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.
  27. 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.
  28. 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.
  29. 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.
  30. 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.
  31. 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.
  32. 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.
  33. 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.
  34. Chakraborty, D.; Rumi, M.A.; Soares, M.J. NK cells, hypoxia and trophoblast cell differentiation. Cell Cycle 2012, 11, 2427–2430.
  35. Faas, M.M.; de Vos, P. Uterine NK cells and macrophages in pregnancy. Placenta 2017, 56, 44–52.
  36. 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.
  37. Mor, G.; Abrahams, V.M. Potential role of macrophages as immunoregulators of pregnancy. Reprod. Biol. Endocrinol. 2003, 1, 119.
  38. 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.
  39. Head, J.R. Uterine natural killer cells during pregnancy in rodents. Nat. Immun. 1996, 15, 7–21.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. Brandes, D.; Anton, E. An electron microscopic cytochemical study of macrophages during uterine involution. J. Cell Biol. 1969, 41, 450–461.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. Sharma, S. Natural killer cells and regulatory T cells in early pregnancy loss. Int. J. Dev. Biol. 2014, 58, 219–229.
  55. 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.
  56. Rosario, G.X.; Ain, R.; Konno, T.; Soares, M.J. Intrauterine fate of invasive trophoblast cells. Placenta 2009, 30, 457–463.
  57. 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.
  58. 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.
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. 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.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. Furukawa, S.; Tsuji, N.; Sugiyama, A. Morphology and physiology of rat placenta for toxicological evaluation. J. Toxicol. Pathol. 2019, 32, 1–17.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. 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.
  84. 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.
  85. 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.
  86. 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.
  87. 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.
  88. 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.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. Akay, M.T.; Kockaya, E.A. The effects of alcohol on rat placenta. Cell Biochem. Funct. 2005, 23, 435–445.
  95. 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.
  96. Estrada-Flores, E.; Yáñez Mendoza, C. Histology of rat placentas treated with methylhydrazine Ro 4-6467. Bol. Estud. Med. Biol. 1990, 38, 59–64.
  97. 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.
  98. Jawerbaum, A.; White, V. Animal models in diabetes and pregnancy. Endocr. Rev. 2010, 31, 680–701.
  99. 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.
  100. 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.
  101. 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.
  102. 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.
  103. 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.
  104. 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.
  105. Fisher, S.J. Why is placentation abnormal in preeclampsia? Am. J. Obstet. Gynecol. 2015, 213, S115–S122.
  106. 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.
  107. Rana, S.; Lemoine, E.; Granger, J.P.; Karumanchi, S.A. Preeclampsia: Pathophysiology, challenges, and perspectives. Circ. Res. 2019, 124, 1094–1112.
  108. 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.
  109. 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.
  110. McLaurin, K.A.; Mactutus, C.F. Polytocus focus: Uterine position effect is dependent upon horn size. Int. J. Dev. Neurosci. 2015, 40, 85–91.
  111. 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.
  112. 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.
  113. 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.
  114. 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.
  115. 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.
  116. 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.
  117. 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.
  118. 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.
  119. Okamoto, K.; Aoki, K. Development of a strain of spontaneously hypertensive rats. Jpn. Circ. J. 1963, 27, 282–293.
  120. 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.
  121. 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.
  122. 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.
  123. 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.
  124. 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.
  125. 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.
  126. 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.
  127. 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.
  128. 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.
  129. 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.
  130. 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.
  131. 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.
  132. 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.
  133. 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.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 307
Revisions: 2 times (View History)
Update Date: 11 May 2022
1000/1000