Introduction
This represents an overview, and not an exhaustive (or systematic literature) review of the use of animal models to study the adverse pregnancy outcomes seen in humans. For several of the outcomes mentioned herein there exist more in-depth reviews and there likely will be more to follow. Nor is this a review of all the data and mechanisms relating to normal and abnormal pregnancy and parturition. I have decided to include a balance between older reports and observations and reviews by revered scientists, as well as newer observations and reviews by seasoned and perhaps less-seasoned investigators. My hope is that clinicians will be able to utilize some of this information to seek out the literature and have more meaningful and profitable discussions with their academic colleagues. I further hope that they will be enticed to engage in regular interactions that will enhance transdisciplinary research in reproductive health. My ultimate agenda is to eliminate the tendency to dismiss work in animal models out of hand because they don’t exactly capture human physiology. In addition, I want to prevent the thinking that little can be learned from observations in humans because of inability to modulate and study specific mechanisms. I would like to see more support for conversations starting from both sides with “This is how I understand how the model behaves and how it might (or not) be reflected in humans. What is your understanding?” I would also like to see the literature, including titles of manuscripts and key words increase visibility of the animal models (e.g. including the words “animal model” and species name) involved in the observations conveyed.
Why animal models?
The limitations of human studies to establish disease causality and of in vivo animal models to replicate human physiology support the use of animal models in an iterative manner. In this process, phenomena described following observational studies in humans drives hypotheses to be tested in animal experiments. Animal experimentation in turn refines hypotheses that can then be tested in humans. This in turn leads to further questions and more productive animal experimentation. In this iterative approach, studies in humans and animals complement each other and can synergize to move our understanding of disease forward. That being said, my bias is that a good animal is not meant to primarily replicate all of what happens in humans, nor is it meant to be directly transferable. A well-working model generates logical and testable hypotheses that are consistent first foremost with existing data in the animal, and possibly in humans as well. The drive for those who primarily use animal models should be to “know thy model”, be able to communicate it effectively to others, and to generate productive integrative and iterative study.
An approach to animal models
In studies in humans, several properties are taken into consideration to determine the appropriateness of the group of patients accessed for a study. These properties may be related to certain demographics or to prevalence of disease. When considering animal models to study adverse pregnancy outcomes, several issues come to mind.
Resources
With decreasing funding through federal and other sources, cost may play a large role in the choice of mode. Larger animal models are likely more costly and research based on these models is receiving less support1. However, certain strains of genetically manipulated mice are also very expensive (http://jaxmice.jax.org). The animal welfare regulatory requirements for non-human primate work are increasingly stringent as is the administrative oversight. Another constraint is the ability to deal with the public relations issues necessary to utilize primate models. Only certain institutions have the capacity, specialized facilities and highly-trained veterinary staff. Depending on the species, there are some zoonotic disease issues which require a very rigorous occupational health program. Another practical issue related to choice of animal models is the presence of experts working with that model. Just as it is often better to watch a relative cooking a family tradition, rather than relying on a recipe, there are likely to be small bits of “inside” or not widely published information about the model that are more easily obtained by direct contact with the investigator utilizing the model.
Placentation
Current thinking would refute the notion that the placenta is just a passive membrane between mother and fetus. Early studies of nutrient uptake suggest that most of the resources delivered to the uterus are utilized by this organ. The placenta is selfish. It is a metabolically hormonally, and immunologically active entity in a triune necessary for successful pregnancy: mother, fetus, placenta. Structurally, the purpose of the placenta in mammals is to bring maternal and fetal circulatory systems in close proximity to facilitate exchange of nutrients, oxygen, waste and other factors2. Several good reviews of comparative placentation exist3–7. Placentae are usually described by the layers existing between fetal trophoblast, which itself envelops fetal vessels and mesenchymal cells, and maternal blood2. The controversy of placentation and the validity of animal models will likely continue because while it is assumed that differences in placentation will lead to different adaptive mechanisms, experimental changing of placentation in certain animals is likely extremely challenging.
The human placenta is said to be hemochorial2, in that maternal blood is in direct contact with fetal trohpblast. There are however, other points of contact between maternal and fetal tissues, for example in the villous structures that anchor the placenta8. The human placenta moreover is said to be interstitial, in that implantation occurs completely within the maternal uterine wall4 thus allowing for multiple points of interaction between maternal and fetal tissues early in gestation. Primates commonly used in research, e.g. baboons, macaque, chimpanzee also have hemochorial placentas3, 6 with more or less invasion upon implantation, and a villous organization, although this is not true for all primates (e.g. lemurs3). The vascular structure of human placenta undergoes a revision in early gestation in which trophblast lines maternal uterine arteries9 to allow for maximal blood flow10. The placenta in rats (see recent review by Soares11) mice and guinea pigs (rodents) is similar to that in humans in that maternal blood is in direct contact with trophoblast. There are subtle(?) structural differences between human and rodent placentae, including the flow of blood due to a labyrinthine as opposed to a villous organization, the depth of trophoblast invasion6, and the trophoblast subpopulations2. For example, an additional layer of trophoblast, the giant cell layer, in addition to cytotrophoblast and syncytital trophoblast has led some authors to call the rodent placenta “hemotrichorial”. Because of only one trophoblast layer, the guinea pig placenta is sometimes referred to as “hemomonochorial”. In addition to structural differences, there are subtle differences in the expression of proteins, such as those involved in immune regulation12–15. While the definitive placenta is in place for a short time relative to gestation in mice and rats2, the longer gestation in guinea pigs makes this less true. Rabbits belong to the group of mammals called lagomorphs. Their placentas are hemochorial with two trophoblast layers, a syncytium and a cytotrophoblast layer which is similar to humans, but organized in a labyrinthine structure2, 5, 16. Sheep and pigs belong to the group of mammals called ruminants (order Artiodactyla) and have a different placental structure where both trophoblast and uterine epithelium are intact but interdigitate (epitheliochorial, think fingers of folded hands) allowing for contact close enough for efficient gas exchange2. In some areas of the sheep placenta, called placentomes there is aggressive interdigitation between trophoblast villi on the fetal side (cotyledon) and the uterus on the maternal side (caruncle) and at points the epithelia form a common syncytium allowing for more efficiency of gas and nutrient exchange. Pigs have a similar but more diffuse placental structure than sheep with less aggressive interdigitation2,17.
Uterine structure, dynamics
The human/primate uterus is a single muscular organ different structurally from the two-horned uterus of rodents (for mice see Margaret J Cook’s book at www.jax.org), pigs18, rabbits16 or sheep19. While the electro-mechanics of the human/primate uterus may be fundamentally different from that seen in other species20, 21, the uteri of rodents22, rabbits23 sheep24 and pigs18 respond to oxytocin, suggesting a common expression of the receptor and most have been used to study the mechanisms underlying uterine contractility in vitro.
Endocrinology of pregnancy
In addition to hormones such as estrogen (discussed elsewhere), progesterone is a key hormone of pregnancy that appears to be differentially regulated in humans and animals25. The particulars of the responsiveness to this hormone and its interaction with estrogen in successful pregnancy remain a topic of intense investigation. In humans, the corpus luteum is the major site of progesterone expression with help from chorionic gonadotropin released by the early conceptus26. Blockade of progesterone during this time causes pregnancy loss26. Major production of progesterone switches to the placenta by 5–6 weeks gestation. Maternal serum levels of progesterone raise post conceptionally and continue to elevate beyond parturition25, 27. However, progesterone has been given with variable success with to treat women with recurrent miscarriage28 and anti-progesterone given late in pregnancy can cause cervical ripening and delivery in some women29 suggesting a complex biology.
Human fetal membranes can produce30 and metabolize progesterone31, and locally produced progesterone metabolites may be important in uterine quiescence and activation32. The human uterus can produce an inhibitory progesterone receptor which increases before parturition33. Finally, progesterone receptor regulation at multiple levels in the cytoplasm and the nucleus may regulate functional progesterone activity leading to parturition34. Progesterone’s regulation during pregnancy in related non-human primates is similar to human pregnancy in several respects including dependence on early production of progesterone by the corpus luteum35, that early pregnancy can be interrupted by antiprogestins36 and that there is not systemic withdrawal before parturition37.
In rodents, the corpus luteum is the source of progesterone that maintains pregnancy. Luteolysis38, removal of the ovaries39, or administration of antiprogestational agents40 leads to uterine activation with increased effective signaling through oxytocin or other receptors and parturition. The difference in serum levels before parturition in mice and rats is said to make these animal a poor model for progesterone regulation in humans. However, further understanding of local progesterone metabolism and responsiveness is likely to reveal mechanisms that are to some extent important in humans and may be a natural stand in for women who do not respond to exogenous progesterone in the prevention of preterm birth. Rats also express an inhibitory receptor that increases in expression before parturition41.
In guinea pigs, in which early pregnancy can be disrupted by antiprogestins42, maternal serum levels, similar to what is seen in humans, rises from the time of conception to a peak in early gestation followed by a transient decrease in late gestation and increasing levels from that point beyond the time of parturition25. Rabbits and sheep in contrast have very low levels of progesterone in the serum as compared to humans, and in these animals, pregnancy brings a slight increase in serum progesterone and a rapid fall before parturition25.
Another important endocrine system related to pregnancy is the hypothalamic pituitary adrenal axis43, both of the mother and the fetus. Activation of the HPA axis by stress or other factors initiates a cascade involving release of corticotropin releasing hormone (CRH) from the hypothalamus, secretion of corticotropin (ACTH) from the anterior pituitary, and action of ACTH on the adrenal to release cortisol and other glucocorticoids which can then exert feedback suppression on their release. This system not only interacts with the immune system, but is also thought to be part of the mechanism underlying poor pregnancy outcomes related to emotional or physiologic stress4445. CRH, a principle mediator of the HPA axis is produced by the placenta and fetal membranes45, and may be a mediator of local estrogen production. In pregnant women, the possibility for multiple sources of increased systemic CRH presents an ongoing challenge in understanding the interaction between maternal stress, fetal stress and normal HPA development in the generation of parturition or preterm birth46. Animal models are likely critical in the examination of this issue in that they can be used to isolate and understand the potential importance of maternal versus fetal HPA, and other factors47, 48 in this process.
In related non-human primates, the placenta also expresses CRH, and development of the fetal adrenal and activation of the fetal HPA axis generate important support signals for normal labor48. Compared to humans, the biochemistry of adrenal steroid production and the development of the fetal adrenal gland in various non-human primates show subtle differences that may need to be considered in choosing a primate model to examine the role of the HPA axis in normal development or prematurity49.
In rats and mice the HPA axis expresses important differences from that found in humans. For example, the major product of HPA axis activation in humans is cortisol, while that in most rodents it is corticosterone50. Moreover, the development of the fetal adrenal gland in rats and mice is markedly different with major relative deficiencies in important enzymes and preference for different substrates. In these species, the response to stress may lead to fundamentally different means of pregnancy failure, including a decreased level of circulating progesterone51. While rodent models may not be ideal for examination of the role of the HPA axis in normal pregnancy, evolving rodent models may be of interest in understanding the interaction of the HPA axis and stress in parental behavior52.
Sheep have been used as a model of maternal53 and fetal HPA axis function during pregnancy. In this animal model, it is the development and activation of the fetal HPA that is the primary driver of parturition54, and stresses such as hypoxia activate the HPA axis in sheep and lead to preterm labor55
Immune system
The maternal-fetal interface in humans includes not only close contact between maternal and fetal cells within the placenta and uterus8 but also within the maternal and fetal circulations, as cellular traffic has been shown in either direction56, 57. The expression of proteins unique to the mother on fetal cells has raised a decades-long debate over the critical pathways and mechanisms needed to assure both immune tolerance and protection of the fetus from infection58. Humans can mount an immune response against fetal antigens during pregnancy59, and it is clear that there is an intricate interaction between maternal immune cells and trophoblast60, 61. This interaction may be of benefit to the evolving conceptus62 or may be involved in early pregnancy loss or other adverse pregnancy outcomes63. Activation of local innate immunity within the myometrium is thought to play a role in parturition64, and in premature uterine contractions65. In humans, certain pathogens are more deleterious during pregnancy as compared to the non pregnant state66 while others are not67 and the role of the placenta as a safe harbor for evolving pathogens has been described68. Some infection syndromes that occur in humans occur only under contrived conditions in animals69. Moreover, some organisms, such as CMV are different in different hosts70. Both the peculiarities of the immune response and the infectious agent must be taken into consideration when using an animal model to understand the function of the immune response during pregnancy.
The maternal- fetal interface in primates expresses similar oligomorphic Major Histocompatibility (MHC) molecules71 and other immune modulating factors48 as are found in humans. Similar populations of immune cells have also been observed in the primate uterus and placenta during pregnancy72–74. Moreover, shared susceptibility to certain infections exists75. In addition, the high degree of sequence similarity between key human and non-human primate protein sequences has supported the use of anti-human antibodies in ELISA and other immune assays to examine the immune response in non-human primates. These factors have made primate models useful for the study of infection, immunity and adverse pregnancy outcome.
Mice have also been used extensively to model both maternal innate and adaptive immunity. There has been extensive study on the trafficking of cells across the maternal-fetal interface76–78 and on the intricate interaction between trophoblast and innate immune cells in gestation79, 80. While there are some differences in the phenotype of natural killer (NK) cells at the maternal-fetal interface81, and differences in the diversity of the MHC molecules expressed on trophoblast subpopulations in humans and mice82, both systems have been used to delineate specific mechanisms and paint a picture of NK cells as “educable”83, 84, supportive of placental structure and development82, but potentially participating in disruption of pregnancy85 (and see below).
The mouse has also been used to examine maternal T cell regulation during pregnancy. As in the human, the pregnant mouse can generate a fetus specific immune response77, including effector and regulatory T cells86, 87. An advantage to the mouse is the ability to vary the genetic difference between mother and fetus. For example, some strains of mice respond to the male antigen, H-Y, and thus maternal immunity can be studied in a situation where mother and fetus are genetically identical, except for the expression of proteins relevant to maleness. The so- called anti H-Y response is generated in mouse pregnancy77, and has been shown to shown modulate both CD488 and CD889 maternal T cells. Several genetically modified antigen systems have been used to examine maternal anti fetal immunity in pregnant mice90. Although human but not mouse T cells can present antigen via MHC II, the mouse has also been used to examine fetal antigen presenting cells during pregnancy91, 92. Integrated studies in mice and humans will likely increase our knowledge of the function of the immune system during pregnancy and reveal the presence and importance of specific pathways.
Guinea pigs and humans have similar immune systems making them a useful tool in the study of relevant human infectious diseases93. Guinea pigs are extensively used in models of anaphylaxis and allergy94. Many tools are now available to examine the immune system in these animals95.
The rabbit has also been used for a variety of immunology and infectious disease research. The whole genome of the rabbit has been sequenced and utilized to determine possible genomic differences in loci responsible for immunity96.
As in humans and mice, systemic immunity during pregnancy has been examined in sheep. Some studies have found no alteration during pregnancy97 while other studies have found the sheep produces pregnancy-specific agents that can suppress immune responses98. In human pregnancy, there is a systemic turnover of a subtype of T cells, bearing gamma and delta chain T cell receptor in the peripheral blood99. These gamma-delta T cells are also present in the deciduas100 and may play a role in fetal protection101. A highly diverse population of gamma delta T cells is present in sheep uterus during pregnancy, providing large numbers of cells for study102, 103. Pigs have also been studied to understand immunity at the maternal- fetal interface, and for example underlined the importance of uterine NK cells104.
Length of gestation and fetal development
In human and other primate gestation, implantation is ~ 7–8 days after ovulation followed by a 10 week long pre-embryonic and embryonic period28. This is followed by a prolonged fetal period resulting in a highly developed fetus in relatively low numbers. During this time multiple insults inside and outside the uterus can disrupt both pregnancy and fetal well being. For ease of experimentation, a shorter length of gestation, such as found in most rodents (i.e. ~ 19–22 days) may be desired. However, the rodent fetus is born less developed than the human105. Currently, tissue-specific inducible promoters, Cre-recombinase and related technology allow for the generation of genetically-based time and tissue-specific modulation of gene expression during mouse pregnancy. These changes can be examined in the developing fetus and the newborn. However this technology may be difficult to obtain, and mice with the desired modifications may not exist. Moreover, the short gestation and small fetal size constrain the ability to make specific surgical or physiologic interventions and relate these to fetal development. While rats are relatively larger, and more amenable to these interventions, the technology to generate targeted gene expression or deletion in rats is less-developed or utilized106.
The guinea pig is a rodent used in many studies of maternal environment and fetal development, as it has a longer gestation of 68 days2, and its offspring are born highly precocious105 with a mature central nervous system at birth105. Another rodent with a longer gestation is the “spiny mouse” of the genus Acomys (not Mus as in mice). This small rodent has a relatively long gestation (38–42 days) and gives birth to a small litter (2–3 pups) that are born highly developed107. These exotic animals however are difficult to manage due to their delicate skin108. There is a long and distinguished history using rabbits to understand early development16 In rabbits, ovulation is induced by mating, resulting in an exactly defined pregnancy and embryonic age assessment. Larger animals, such as sheep, have long and lower order gestations (singleton/twin) and produce highly developed offspring and thus have been used for studies of pregnancy insult on fetal development109.
Preeclampsia
Preeclampsia is a pregnancy-related syndrome that affects multiple systems and clinically presents as hypertension, proteinuria, edema and in its more sever forms evidence of fetal compromise, neurologic abnormality, liver and hematologic dysfunction110. The complexity of the syndrome defies the development of a panel of genetic screens or biomarkers111. While the basic cause of the disease is as yet unknown, multiple hypotheses exist. These include failure of placentation112 and thus reduced utero-placental perfusion, intolerance to volume expansion generated by pregnancy113, infection114 and inflammation115. It is hotly debated as to whether failed placentation is caused or a by-product of broken maternal immune tolerance116, 117. Many agree that a common final pathway to the manifestation of the disease is endothelial cell damage occurring in a variety of vascular beds118.
While the disease is thought of as being uniquely human, many recognize the potential positive role of integration of research in human and animal models in understanding the underlying mechanisms119, 120. The hallmarks of preeclampsia most sought after in animal models are hypertension, renal dysfunction (proteinuria), and further, conditions such as poor trophoblast invasion and endothelial damage. Current models address some of these issues.
There have been rare reports of spontaneous preeclampsia in related non-human primates121. These species have also been used to develop models of pregnancy-related hypertension and proteinuria based on injection during mid- gestation of inflammatory mediators, such as Tumor Necrosis Factor122 or antibodies to interleukin 10123.
There are strains of mice that spontaneously develop hypertension, proteinuria, smaller litters and fetal demise and these have been used to model preeclampsia124, 125. There are also models of spontaneous pregnancy-associated hypertension with fetal compromise in rats126. There also exist genetically manipulated mouse and rat models. In one interesting genetic model of hypertension in pregnancy, female mice transgenic for human angiotensinogen are mated to males transgenic for human rennin127. The resulting pregnancy is marked by distortion of placental anatomy, elevation of circulation Vascular Endothelial Growth Factor (VEGF) receptor in mid gestation (12–13 of 19–20 days), hypertension, fetal intrauterine growth retardation and systemic maternal disorders including proteinuria and convulsion. In the rat version of this model128 the hypertensive disease experienced by the pregnant rat is thought related to secretion of rennin from the placenta into the maternal circulation129. Interestingly, the extent of trophoblast invasion into the spiral arteries in these pregnancies was increased compared with non pregnant animals, and the breeding, when done in reverse (dams transgenic for rennin males for angiontensinogen) was associated with lower blood pressure. Overall studies in humans, in vitro, and in animal models have yielded interesting hypotheses surrounding the placenta as a independent factor in the development of preeclampsia. Animal models, in conjunction with genetic studies in humans113 will likely elucidate an important underlying mechanism(s) for the disease.
To model the presumed decrease in placental perfusion that occurs as part of the mechanism proposed to incite preeclampsia130, workers have ligated various levels of the uterine artery. The RUPP or reduced uterine perfusion pressure model (reviewed in131) is performed in rats and several other animals. In rats the model is performed at around 14 days of gestation by placing a clip above the aortic bifurcation and on both sides of the uterine arcade to prevent utero-ovarian collateral flow. This results in a 40% or more reduction in flow to the developing fetal-placental units, and the resulting disease includes hypertension, renal damage (proteniuria), increased vascular reactivity and small pups. In rats an alternative of this model is based on increased salt intake and administration of desoxycorticosterone acetate132, which generates hypertension, convulsions, proteinuria and renal lesions133.
Other rodent models of reduced vascular function have utilized injection of inhibitors of nitric oxide (i.e. L-NAME (N-omega-nitro-l-arginine methyl ester134), or over expression of soluble VEGF receptor (sVEGFRI, sFLT1) or members of the Transforming Growth Factor β receptor complex (i.e. endoglin). Adenovirus–driven over expression of sFLT1 in pregnant rats leads to hypertension and proteinuria in a dose-dependent manner135, and this is enhanced by over expression of soluble endoglin136.
Other animals have also been used to develop models of preeclampsia. In guinea pigs there have been reports of a naturally occurring preeclampsia-like syndrome137. In addition, it has been observed that banding of the uterine arteries as well as transaction of the ovarian arteries before pregnancy results in later pregnancy hypertension, proteinuria and elevated creatinine138. Moreover, early observations of constriction of the aorta in pregnant rabbits revealed that such manipulation generated hypertension, proteinuria, weight gain, and reduced weight of the fetus139. Finally, sheep experience what is call toxemia of pregnancy, that appears to be a very different metabolic disorder as compared to preeclampsia140, but does include proteinuria and inflammation141.
Intrauterine growth restriction
In humans, intrauterine growth restriction (IUGR) can be an independent outcome of a fetal abnormality or related to placental insufficiency due to a number of maternal/environmental factors including poor nutrition, smoking and chronic infection, or in the context of preeclampsia142. Poor intrauterine growth has been extensively studied in animals143, and thus the time is ripe for more extensive integration of the information in humans and animals.
In related primates, IUGR has been induced using various levels of maternal nutrient restriction144, and surgical manipulation of placental blood supply145 among other interventions. In animals with litters, there is evidence that the fetuses placed at a distance from the main uterine artery are smaller146. In pigs, a proportion of piglets in a litter is naturally small146, 147.
In mice, genetic models of deficiency in key molecules such as eNOS have been generated and pups of these pregnancies show IUGR148 while their mothers do not show a characteristic mid-gestation drop in systemic blood pressure149. In mice and rats bilateral uterine artery ligation late in gestation leads to fetal intrauterine growth retardation, neurologic deficiency and metabolic derangement150. Uterine artery ligation at mid gestation (~day 30 of 70) in guinea pigs also produces growth restriction151.
Ligation of utero-placental vessels in rabbits on day 25 of a 31 day gestation produces small pups that show deficiencies in neurobehavioral development152. Administration of L-NAME on day 24–28 of gestation is also used to model IUGR in a rabbits and this model results in growth retarded fetuses and decreased flow, as determined by 3D power Doppler Angiography, in each utero-placental unit153.
In sheep, there are several models of fetal growth restriction109. These include maternal calorie restriction154 emobilization of the umbilico-placental arteries155, and disruption of the uterine epithelium in close contact with trophoblast in the placenta156. Maternal hyperthermia gestation day 35–40/~147 gestation157–159 has been shown to produce asymmetrical growth restriction and decreased placental mass, and abnormal umbilical arterial and aortic Doppler velocimetry160, while placement of the mother in hypoxic conditions also limits fetal growth161. Some breeds of sheep are more amenable to these manipulations than others109, suggesting that with advanced technology and genome sequencing, these animals may be used to examine gene-gene- and gene-environment interaction in the development of this disease.
Recurrent miscarriage
Human pregnancy is less efficient than many other species, as nearly 50% of conceptions fail28. In humans, recurrent miscarriage is a complex syndrome that likely incorporates several types of defects in genetics, implantation, placentation, metabolism, and hormonal support of the conceptus28, 162 or stress163. Thoroughbred horses164 and commercial pork breeds165 also have a high rate of spontaneous abortion. One idea that drives the field is that disregulation of maternal innate or adaptive immunity initiates or contributes significantly to the disease166, 167. Immune modulation as a treatment in human disease has met with variable success, and this is still a matter of controversy168. Whether an initial metabolic, structural or related defect leads to immune activation and a subsequent deleterious response or an initial loss of immune regulation leads directly to tissue disregulation and destruction is still a matter of debate in some circles. Thus, the issue of immune-mediated recurrent pregnancy loss is one that is likely amenable to iterative studies in animal models and humans.
In primates, parental sharing of MHC has been correlated with decreased pregnancy success169. Moreover, administration of anti-progestational agents can produce early pregnancy loss, as in humans170. Primates have also been used to develop models of pregnancy loss related to infections171.
A well-known mouse model of pregnancy loss involves the breeding of a CBA strain female mouse with a male DBA strain male. Depending on the source and housing (level of pathogens present) of the mice, pregnancies can be affected by high levels of fetal-placental degeneration (referred to as “resorption”)172 and infiltration with NK and other immune cells173. In this model, resorption of the fetuses occurs at approximately day 9–12 of gestation174. Contributors to increased fetal loss in this model include stress175, inflammation176, 177 abnormal cytokine milieu within the placenta/decidua178, 179, disrupted regulatory immune modulation180, 181 and abnormal placental vascular development182, 183. Several methods of immune modulation184–187 have been shown to decrease fetal loss in this model, but few if any have been successfully translated to clinical care28. More recent models of pregnancy loss in mice involves chemically targeting86 depletion87 or genetic deficiency of a subpopulation188 of regulatory T cells in normal C57Bl/6 females mated to same strain or allogeneic males. An alternative immune- based models of pregnancy loss involved NK T cell activation in certain strains of mice189, and systemic immune activation leading to ovarian insufficiency190.
Study of the high rate of pregnancy loss in commercial pork breeds has further suggested the role of immune cells in supporting successful pregnancy191. Moreover, Guinea pigs (for example192) and Sheep193 have been used in models of early pregnancy loss in response to infection. Finally, autoimmune related loss, as in the antiphospholipid syndrome has been modeled in rabbits194.
Preterm birth/prematurity
The study of premature birth presents at least three major issues that are amenable to studies in animal models195. The first is the discovery of mechanisms leading to premature labor. A second pertains to delineating consequences of being born premature. Thirdly, animal models have been employed to devise ways to better manage the premature neonate. While the factors contributing to prematurity in humans are far from understood, emerging data suggests that preterm births fall into definable categories196. These categories include preterm births in women who have a history of preterm birth197, in women with multiple gestations198, women who are undergoing an infectious or inflammatory process199–201, women who undergo social and emotional stress202, and women who have medically indicated or physician-driven premature births203. While in general, animals are not said to experience preterm birth, there is variability in gestation within species. Recent data for example, suggests that there is significant variability in mouse gestation related to strain204 or cytokine expression205.
Endocrine disruption
Progesterone has been used in various formats for the prevention of preterm birth206, 207. Clearly, there are patients who respond to progesterone, and those who do not. Only a proportion of women respond to vaginal progesterone, particularly if the cervix in shortened. Even amongst women with a tendency towards preterm birth as evidenced by a previous premature delivery, there are those who respond to regular administration of a progestational agent while others do not. Finally, with the reinstatement of progesterone and related agents in the past decade, there remains a significant incidence of preterm birth208. Use of animal models in conjunction with a more careful study of responders versus non responders209 in human trials of progesterone and related agents will enhance our understanding and management of pregnancy.
Decreased relative progesterone activity can be modeled in mice via oophorectomy or administration of agents such as RU486 in primates (see above). Preterm birth can also be generated in rabbits using RU486210. Novel models of endocrine disruption in mice211and likely other animals are being developed. In several animal models, a signal from the fetus, the placenta, or the endometrium leads directly or indirectly through a systemic response circuit to decreased relative progesterone activity and increased estrogen activity212, 213. This in turn leads to increased prostaglandin (increased production, decreased hydrolysis), uterine contractions, cervical ripening and subsequent rupture of membranes and expulsion of the fetus. For example, the stress response, thought to be mediated by cortisol is modeled in sheep by systemic administration of glucocorticoid214 or in the fetus215. The complexity of these models is likely to increase, and bring forth possible means to modify the process of disrupted endocrine function in premature birth34.
Immune/inflammatory In very well studied models in mice (for examples216–218), rabbits219,220, 221and primates222–224 exposure of the uterus to an inflammatory signal or infectious process leads to an increased local presence of inflammatory cells218, 225 and feeds into the mechanisms resulting in increased uterine contractions or cervical ripening and subsequent preterm birth. An interesting alternative inflammatory model involves injection of a major lung surfactant protein which is thought to activate uterine macrophages and lead to preterm birth213. Guinea pig uterus is particularly sensitive to mast cell secreted mediators, making this a potentially important model for examining the role of allergy an preterm birth226,227.
A salient example of the iterative nature of successful research in animals and humans is the work surrounding Toll-like receptors and preterm birth. In the early 1960s, it was recognized that urinary tract infections in women were associated with preterm birth228, 229. The 1970’s brought forth reports that lippopysaccharide, a component of the outer membrane of gram negative bacteria interrupts early and late pregnancy in mice230 and rats231. In 1985, the Toll gene in Drosophila was cloned232. The early 90’s brought studies suggesting that LPS-induced preterm delivery induced changes in local and systemic cytokines including tumor necrosis factor-alpha and interleukins 1,6, and 8233, 234. In the late 90’s, the drosophila Toll gene was linked to antifungal immunity and the delineation of the Toll-like receptor (TLR) family of proteins began235–237. At this time it was recognized that a certain strain of mice was hypo-responsive to LPS238. That these mice possessed mutations in the Tlr4 locus generated much excitement that Tlr4 was the innate receptor for LPS and the link between infection and LPS-mediated inflammation. The early 2000s brought studies trying to link polymorphisms in Tlr4 to LPS responsiveness, preterm labor, and preterm premature rupture of membranes in humans239. In the mid-late 2000s, investigators using mouse models determined that preterm delivery induced by bacteria expressing LPS is dependent on TLR4 signaling.240 They delineated several relevant pathway constituents, including Myeloid Differentiation primary-response gene 88 (MyD88)241, nuclear factor kappa B(NFκB)242 cytokines, such as tumor necrosis factor and others243 and prostaglandins244. At about this time began studies of expression and regulation of these molecules and their pathways in human placenta, uterus and decidua245, 246 and the correlation between Tlr4 expression and other adverse pregnancy outcomes in humans115, 247. Recently, a TLR4 antagonist was tested in a rhesus model for decreasing LPS-induced inflammation and uterine contractions223. Moreover, the role of other TLR molecules in preterm birth248–250 has generated experiments linking bacterial and viral co-infection with preterm birth251, suggesting synergy in signaling from two TLRs. Finally, data are developing that link circulating fetal DNA and yet other TLRS with this process252.
Important complications of prematurity in humans that are investigated in animal models include white-mater damage and cerebral hemorrhage which is thought to be the basis for cerebral palsy and learning disability253. Studies of preterm birth in humans have supported the idea that not only infection but also inflammation is a significant underlying cause of preterm birth254. In addition, this data has contributed to the idea that the fetus generates a significant inflammatory response under these conditions255 and that this response may subject the fetal brain to processes leading to cerebral palsy256. Several animal models have been used to examine fetal neurologic insult in the context of maternal systemic infection or inflammation and the resulting preterm labor. These studies have included systemic injection of LPS in pregnant sheep257 and intrauterine injection in rabbits258 and in mice259–261. The mouse model of preterm birth initiated with injection of LPS revealed the important role of the cytokine interleukin 10262, 263. In addition, human studies have suggested the potential role of this cytokine in modifying preterm birth related brain injury264. The study of inflammation-related preterm birth and brain injury offers another opportunity for productive iterative study in humans and animals.
Adverse fetal programming
“Programming” is said to occur during “a critical period when the system is plastic and sensitive to the environment followed by loss of plasticity and a fixed functional capacity”265. “Fetal programming” in humans is said to occur as a result of adaptation to undernutrition in an adverse intrauterine environment contributes significantly to obesity, metabolic syndrome, and cardiovascular disease266. Increasingly, animal models are being used to delineate these mechanisms, and several models utilizing rats, mice, rabbits sheep, and nonhuman primates have been utilized (see Fischer16, Seki267, and Vuguin158 for reviews)]. Some of these models proceed through well recognized defects in fetal development, such as IUGR. This issue is one that is ripe for an iterative process involving studies in animals and humans. An area that would be particularly amenable to animal experimentation would be the examination of multigenerational effects of exposure during pregnancy268.
Is the future now? Bioinformatics and the iterative use of animal models
Although the relevant tissue in humans is sometime hard to access, genetic variability found from sampling peripheral blood can be informative in conjunction with specific gene manipulation in rodents. For example, technology exists to manipulate embryos by using viral constructs to target genes to trophoblast11, 269. It is therefore not difficult to imagine an experimental paradigm whereby candidate genes from human genetic studies would be considered for over expression or “knock down” in trophoblast using this technology. Pregnancies using these manipulated embryos could then be observed or further challenged and observed for preterm birth. In this way, and perhaps many others, bioinformatics, systems biology and the use of animal models could be woven into and increasingly efficient iterative method to understand the complex biology of abnormal pregnancy.
The overwhelming increase in genomic, transcriptomic, proteomic, metabolomic, and now microbiomic data in human disease requires continued development of methodologies to probe and understand existing data. Once understood, however, specific genes/proteins reveal themselves as important and these can then be analyzed in animal models270. Similarly, “omic” data from animal models can theoretically be used to query existing repositories from human studies271.
Finally, the large amount of data in both humans and animal will further advance our ability to mathematically model pregnancy272 and perform in silico experiments and use machine learning273. The time may come when the iterative method I propose between human studies and animal models may require this third facet in the quest to understand reproduction.
Post note
This shallow overview was meant to increase curiosity and enhance discussion between clinicians and researchers who utilize animal models in the study of adverse reproductive outcomes. The solution to these problems will come from an integrative and iterative method that starts from clear identification of studies in animals in the literature, an enhanced understanding of the available models and the increased willingness to see value in what at first may seem obscure.
Acknowledgements
I apologize to those colleagues whose excellent work was, due to space considerations, not cited herein. I am grateful for present and past support from the Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, NIH RO1 HD043185, and The March of Dimes Prematurity Research Initiative. I am also grateful for the intellectual support of my colleagues in The Preterm Birth International Collaborative (PREBIC).
References
- 1.Roberts RM, Smith GW, Bazer FW, Cibelli J, Seidel GE, Bauman DE, Reynolds LP, Ireland JJ. Farm Animal Research in Crisis. Science. 2009;324:468–469. doi: 10.1126/science.1168521. [DOI] [PubMed] [Google Scholar]
- 2.Enders AC, Blankenship TN. Comparative placental structure. Adv Drug Deliv Rev. 1999;38:3–15. doi: 10.1016/s0169-409x(99)00003-4. [DOI] [PubMed] [Google Scholar]
- 3.Wildman DE, Chen C, Erez O, Grossman LI, Goodman M, Romero R. Evolution of the mammalian placenta revealed by phylogenetic analysis. Proc Natl Acad Sci U S A. 2006;103:3203–3208. doi: 10.1073/pnas.0511344103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.James JL, Carter AM, Chamley LW. Human placentation from nidation to 5 weeks of gestation. Part II: Tools to model the crucial first days. Placenta. 2012;33:335–342. doi: 10.1016/j.placenta.2012.01.019. [DOI] [PubMed] [Google Scholar]
- 5.Enders AC, Carter AM. Review: The evolving placenta: Different developmental paths to a hemochorial relationship. Placenta. 2012;33(Supplement):S92–S98. doi: 10.1016/j.placenta.2011.10.009. [DOI] [PubMed] [Google Scholar]
- 6.Carter AM. Animal Models of Human Placentation – A Review. Placenta. 2007;28(Supplement):S41–S47. doi: 10.1016/j.placenta.2006.11.002. [DOI] [PubMed] [Google Scholar]
- 7.Carter AM, Pijnenborg R. Evolution of invasive placentation with special reference to non-human primates. Best Practice & Research Clinical Obstetrics & Gynaecology. 2011;25:249–257. doi: 10.1016/j.bpobgyn.2010.10.010. [DOI] [PubMed] [Google Scholar]
- 8.Benirschke K. Remarkable placenta. Clin Anat. 1998;11:194–205. doi: 10.1002/(SICI)1098-2353(1998)11:3<194::AID-CA8>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
- 9.Fisher SJ, Cui TY, Zhang L, Hartman L, Grahl K, Zhang GY, Tarpey J, Damsky CH. Adhesive and degradative properties of human placental cytotrophoblast cells in vitro. The Journal of Cell Biology. 1989;109:891–902. doi: 10.1083/jcb.109.2.891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Pijnenborg R, Vercruysse L, Brosens I. Deep placentation. Best Pract Res Clin Obstet Gynaecol. 2011;25:273–285. doi: 10.1016/j.bpobgyn.2010.10.009. [DOI] [PubMed] [Google Scholar]
- 11.Soares MJ, Chakraborty D, Karim Rumi MA, Konno T, Renaud SJ. Rat placentation: An experimental model for investigating the hemochorial maternal-fetal interface. Placenta. 2012;33:233–243. doi: 10.1016/j.placenta.2011.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Redline RW, Lu CY. Localization of Fetal Major Histocompatiblity Complex antigens and Maternal Leukocytes in Murine Placenta. Lab Invest. 1989;61(1):27–36. [PubMed] [Google Scholar]
- 13.King A, Boocock C, Sharkey MA, Gardner L, Beretta A, Siccardi AG, Loke YW. Evidence for the expression of HLA-C class I mRNA and protein by human first trimester trophoblast. JImmunol. 1996;156:2068–2076. [PubMed] [Google Scholar]
- 14.Kovats S, Main EK, Librach C, Stubblebine M, Fisher SJ, DeMars R. A Class I Antigen HLA-G, Expressed in Human Trophoblasts. Science. 1990;248:220–223. doi: 10.1126/science.2326636. [DOI] [PubMed] [Google Scholar]
- 15.Kruse A, Hallmann R, Butcher EC. Specialized patterns of vascular differentiation antigens in the pregnant mouse uterus and the placenta. BiolReproduction. 2000;61(6):1393–1401. doi: 10.1095/biolreprod61.6.1393. [DOI] [PubMed] [Google Scholar]
- 16.Fischer B, Chavatte-Palmer P, Viebahn C, Navarrete Santos A, Duranthon V. Rabbit as a reproductive model for human health. Reproduction. 2012;144:1–10. doi: 10.1530/REP-12-0091. [DOI] [PubMed] [Google Scholar]
- 17.Amoroso EC. The evolution of viviparity. Proc R Soc Med. 1968;61:1188–1200. [PMC free article] [PubMed] [Google Scholar]
- 18.Dittrich R, Mueller A, Oppelt PG, Hoffmann I, Beckmann MW, Maltaris T. Differences in muscarinic-receptor agonist–, oxytocin-, and prostaglandin-induced uterine contractions. Fertility and Sterility. 2009;92:1694–1700. doi: 10.1016/j.fertnstert.2008.08.117. [DOI] [PubMed] [Google Scholar]
- 19.Hadek R. Histochemical studies on the uterus of the sheep. American Journal of Veterinary Research. 1958;19:882–886. [PubMed] [Google Scholar]
- 20.Young RC. Myocytes, myometrium, and uterine contractions. Ann N Y Acad Sci. 2007;1101:72–84. doi: 10.1196/annals.1389.038. [DOI] [PubMed] [Google Scholar]
- 21.Young RC, Goloman G. Mechanotransduction in rat myometrium: coordination of contractions of electrically and chemically isolated tissues. Reproductive Sciences. 2011;18:64–69. doi: 10.1177/1933719110379637. [DOI] [PubMed] [Google Scholar]
- 22.Beretsos P, Loutradis D, Koussoulakos S, Margaritis LH, Kiapekou E, Mastorakos G, Papaspirou I, Makris N, Makrigiannakis A, Antsaklis A. Oxytocin receptor is differentially expressed in mouse endometrium and embryo during blastocyst implantation. Ann N Y Acad Sci. 2006;1092:466–479. doi: 10.1196/annals.1365.046. [DOI] [PubMed] [Google Scholar]
- 23.Cross BA. ON THE MECHANISM OF LABOUR IN THE RABBIT. J Endocrinol. 1958;16:261–276. doi: 10.1677/joe.0.0160261. [DOI] [PubMed] [Google Scholar]
- 24.Sheldrick EL, Flint APF. Endocrine control of uterine oxytocin receptors in the ewe. J Endocrinol. 1985;106:249–258. doi: 10.1677/joe.0.1060249. [DOI] [PubMed] [Google Scholar]
- 25.Mitchell BF, Taggart MJ. Are animal models relevant to key aspects of human parturition? American Journal of Physiology - Regulatory, Integrative and Comparative Physiology. 2009;297:R525–R545. doi: 10.1152/ajpregu.00153.2009. [DOI] [PubMed] [Google Scholar]
- 26.Norwitz ER, Schust DJ, Fisher SJ. Implantation and the Survival of Early Pregnancy. New England Journal of Medicine. 2001;345:1400–1408. doi: 10.1056/NEJMra000763. [DOI] [PubMed] [Google Scholar]
- 27.Tulchinsky D, Hobel CJ, Yeager E, Marshall JR. Plasma estrone, estradiol, estriol, progesterone, and 17-hydroxyprogesterone in human pregnancy. I. Normal pregnancy. Am J Obstet Gynecol. 1972;112:1095–1100. doi: 10.1016/0002-9378(72)90185-8. [DOI] [PubMed] [Google Scholar]
- 28.Porter TF, Scott JR. Evidence-based care of recurrent miscarriage. Best Practice & Research Clinical Obstetrics & Gynaecology. 2005;19:85–101. doi: 10.1016/j.bpobgyn.2004.11.005. [DOI] [PubMed] [Google Scholar]
- 29.Neilson JP. Mifepristone for induction of labour. Cochrane Database Syst Rev. 2000;4:CD002865. doi: 10.1002/14651858.CD002865. [DOI] [PubMed] [Google Scholar]
- 30.Maslar IA, Hess DL, Buckmaster JG, Lazur JJ, Stanczyk FZ, Novy MJ. Steroid production by early pregnancy human placental villi in culture. Placenta. 1990;11:277–288. doi: 10.1016/s0143-4004(05)80274-7. [DOI] [PubMed] [Google Scholar]
- 31.Milewich L, Gant NF, Schwarz BE, Chen GT, Macdonald PC. Initiation of human parturition. IX. Progesterone metabolism by placentas of early and late human gestation. Obstet Gynecol. 1978;51:278–280. [PubMed] [Google Scholar]
- 32.Sheehan PM. A possible role for progesterone metabolites in human parturition. Aust N Z J Obstet Gynaecol. 2006;46:159–163. doi: 10.1111/j.1479-828X.2006.00548.x. [DOI] [PubMed] [Google Scholar]
- 33.Mesiano S, Chan E-C, Fitter JT, Kwek K, Yeo G, Smith R. Progesterone Withdrawal and Estrogen Activation in Human Parturition Are Coordinated by Progesterone Receptor A Expression in the Myometrium. Journal of Clinical Endocrinology & Metabolism. 2002;87:2924–2930. doi: 10.1210/jcem.87.6.8609. [DOI] [PubMed] [Google Scholar]
- 34.Williams KC, Renthal NE, Condon JC, Gerard RD, Mendelson CR. MicroRNA-200a serves a key role in the decline of progesterone receptor function leading to term and preterm labor. Proc Natl Acad Sci U S A. 2012;109:7529–7534. doi: 10.1073/pnas.1200650109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Duffy DM, Stouffer RL, Stewart DR. Dissociation of relaxin and progesterone secretion from the primate corpus luteum by acute administration of a 3 beta-hydroxysteroid dehydrogenase inhibitor during the menstrual cycle. Biology of Reproduction. 1995;53:447–453. doi: 10.1095/biolreprod53.2.447. [DOI] [PubMed] [Google Scholar]
- 36.Micks E, Shekell T, Stanley J, Zelinski M, Martin L, Riefenberg S, Adevai T, Jensen J. Medical termination of pregnancy in cynomolgus macaques. J Med Primatol. 2012;41:394–402. doi: 10.1111/jmp.12019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Walsh SW, Stanczyk FZ, Novy MJ. Daily hormonal changes in the maternal, fetal, and amniotic fluid compartments before parturition in a primate species. J Clin Endocrinol Metab. 1984;58:629–639. doi: 10.1210/jcem-58-4-629. [DOI] [PubMed] [Google Scholar]
- 38.Erlebacher A, Zhang D, Parlow AF, Glimcher LH. Ovarian insufficiency and early pregnancy loss induced by activation of the innate immune system. The Journal of Clinical Investigation. 2004;114:39–48. doi: 10.1172/JCI20645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Terakawa J, Watanabe T, Obara R, Sugiyama M, Inoue N, Ohmori Y, Hosaka YZ, Hondo E. The complete control of murine pregnancy from embryo implantation to parturition. Reproduction. 2012;143:411–415. doi: 10.1530/REP-11-0288. [DOI] [PubMed] [Google Scholar]
- 40.Dao B, Vanage G, Li XJ, Bardin CW, Koide SS. Comparative effectiveness of three antiprogestins alone and in combination with anordiol in terminating pregnancy in the rat. Contraception. 1997;55:35–40. doi: 10.1016/s0010-7824(96)00239-9. [DOI] [PubMed] [Google Scholar]
- 41.Fang X, Wong S, Mitchell BF. Messenger RNA for progesterone receptor isoforms in the late-gestation rat uterus. American Journal of Physiology - Endocrinology and Metabolism. 2002;283:E1167–E1172. doi: 10.1152/ajpendo.00116.2002. [DOI] [PubMed] [Google Scholar]
- 42.Elger W, Fahnrich M, Beier S, Qing SS, Chwalisz K. Endometrial and myometrial effects of progesterone antagonists in pregnant guinea pigs. Am J Obstet Gynecol. 1987;157:1065–1074. doi: 10.1016/s0002-9378(87)80134-5. [DOI] [PubMed] [Google Scholar]
- 43.Mastorakos G, Ilias I. Maternal and fetal hypothalamic-pituitary-adrenal axes during pregnancy and postpartum. Annals of the New York Academy of Sciences. 2003;997:136–149. doi: 10.1196/annals.1290.016. [DOI] [PubMed] [Google Scholar]
- 44.Entringer S, Buss C, Andersen J, Chicz-DeMet A, Wadhwa PD. Ecological momentary assessment of maternal cortisol profiles over a multiple-day period predicts the length of human gestation. Psychosom Med. 2011;73:469–474. doi: 10.1097/PSY.0b013e31821fbf9a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Torricelli M, Novembri R, Bloise E, De Bonis M, Challis JR, Petraglia F. Changes in placental CRH, urocortins, and CRH-receptor mRNA expression associated with preterm delivery and chorioamnionitis. Journal of Clinical Endocrinology & Metabolism. 2011;96:534–540. doi: 10.1210/jc.2010-1740. [DOI] [PubMed] [Google Scholar]
- 46.Smith R, Smith JI, Shen X, Engel PJ, Bowman ME, McGrath SA, Bisits AM, McElduff P, Giles WB, Smith DW. Patterns of plasma corticotropin-releasing hormone, progesterone, estradiol, and estriol change and the onset of human labor. J Clin Endocrinol Metab. 2009;94:2066–2074. doi: 10.1210/jc.2008-2257. [DOI] [PubMed] [Google Scholar]
- 47.Pearce BD, Grove J, Bonney EA, Bliwise N, Dudley DJ, Schendel DE, Thorsen P. Interrelationship of cytokines, hypothalamic-pituitary-adrenal axis hormones, and psychosocial variables in the prediction of preterm birth. Gynecol Obstet Invest. 2010;70:40–46. doi: 10.1159/000284949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gravett MG, Novy MJ. Endocrine-immune interactions in pregnant non-human primates with intrauterine infection. Infectious Diseases in Obstetrics and Gynecology. 1997;5:142–153. doi: 10.1155/S1064744997000227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Conley AJ, Pattison JC, Bird IM. Variations in adrenal androgen production among (nonhuman) primates. [Review] [171 refs] Seminars in Reproductive Medicine. 2004;22:311–326. doi: 10.1055/s-2004-861548. [DOI] [PubMed] [Google Scholar]
- 50.Ogunsua AO, de Nicola AF, Traikov H, Birmingham MK, Levine S. Adrenal steroid biosynthesis by different species of mouselike rodents. Gen Comp Endocrinol. 1971;16:192–199. doi: 10.1016/0016-6480(71)90031-1. [DOI] [PubMed] [Google Scholar]
- 51.Parker VJ, Menzies JR, Douglas AJ. Differential changes in the hypothalamic-pituitary-adrenal axis and prolactin responses to stress in early pregnant mice. J Neuroendocrinol. 2011;23:1066–1078. doi: 10.1111/j.1365-2826.2011.02204.x. [DOI] [PubMed] [Google Scholar]
- 52.Harris BN, Saltzman W, de Jong TR, Milnes MR. Hypothalamic-pituitary-adrenal (HPA) axis function in the California mouse (Peromyscus californicus): Changes in baseline activity, reactivity, and fecal excretion of glucocorticoids across the diurnal cycle. Gen Comp Endocrinol. 2012;179:436–450. doi: 10.1016/j.ygcen.2012.08.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Keller-Wood M, Cudd TA, Norman W, Caldwell SM, Wood CE. Sheep model for study of maternal adrenal gland function during pregnancy. Lab Anim Sci. 1998;48:507–512. [PubMed] [Google Scholar]
- 54.Bernal AL. Mechanisms of labour—biochemical aspects. BJOG: An International Journal of Obstetrics & Gynaecology. 2003;110:39–45. doi: 10.1046/j.1471-0528.2003.00023.x. [DOI] [PubMed] [Google Scholar]
- 55.Myers DA, Ducsay CA. Adrenocortical and adipose responses to high-altitude-induced, long-term hypoxia in the ovine fetus. J Pregnancy. 2012;2012:681306. doi: 10.1155/2012/681306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bianchi DW, Zickwolf GK, Yih MC, Flint A, Geifman OH, Erikson MS, Williams JM. Erythroid specific antibodies enhance detection of fetal nucleated erythrocytes in maternal blood. Prenatal Diagnosis. 1993;13:293–300. doi: 10.1002/pd.1970130408. [DOI] [PubMed] [Google Scholar]
- 57.Adams KM, Nelson JL. Microchimerism: An Investigative Frontier in Autoimmunity and Transplantation. JAMA. 2004;291:1127–1131. doi: 10.1001/jama.291.9.1127. [DOI] [PubMed] [Google Scholar]
- 58.Bonney EA, Matzinger P. Much IDO about pregnancy. Nat Med. 1998;4:1128–1129. doi: 10.1038/2624. [DOI] [PubMed] [Google Scholar]
- 59.Lissauer D, Piper K, Goodyear O, Kilby MD, Moss PA. Fetal-specific CD8+ cytotoxic T cell responses develop during normal human pregnancy and exhibit broad functional capacity. Journal of Immunology. 2012;189:1072–1080. doi: 10.4049/jimmunol.1200544. [DOI] [PubMed] [Google Scholar]
- 60.Lash GE, Naruse K, Robson A, Innes BA, Searle RF, Robson SC, Bulmer JN. Interaction between uterine natural killer cells and extravillous trophoblast cells: effect on cytokine and angiogenic growth factor production. Hum Reprod. 2011;26:2289–2295. doi: 10.1093/humrep/der198. [DOI] [PubMed] [Google Scholar]
- 61.Huppertz B, Berghold VM, Kawaguchi R, Gauster M. A variety of opportunities for immune interactions during trophoblast development and invasion. Am J Reprod Immunol. 2012;67:349–357. doi: 10.1111/j.1600-0897.2012.01124.x. [DOI] [PubMed] [Google Scholar]
- 62.Hanna N, Bonifacio L, Weinberger B, Reddy P, Murphy S, Romero R, Sharma S. Evidence for interleukin-10-mediated inhibition of cyclo- oxygenase-2 expression and prostaglandin production in preterm human placenta. Am J Reprod Immunol. 2006;55:19–27. doi: 10.1111/j.1600-0897.2005.00342.x. [DOI] [PubMed] [Google Scholar]
- 63.Fukui A, Funamizu A, Yokota M, Yamada K, Nakamua R, Fukuhara R, Kimura H, Mizunuma H. Uterine and circulating natural killer cells and their roles in women with recurrent pregnancy loss, implantation failure and preeclampsia. Journal of Reproductive Immunology. 2011;90:105–110. doi: 10.1016/j.jri.2011.04.006. [DOI] [PubMed] [Google Scholar]
- 64.Shynlova O, Nedd-Roderique T, Li Y, Dorogin A, Lye SJ. Myometrial immune cells contribute to term parturition, preterm labour and post-partum involution in mice. J Cell Mol Med. 2012;4:1582–4934. doi: 10.1111/j.1582-4934.2012.01650.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gravett MG, Witkin SS, Haluska GJ, Edwards JL, Cook MJ, Novy MJ. An experimental model for intraamniotic infection and preterm labor in rhesus monkeys. American Journal of Obstetrics & Gynecology. 1994;171:1660–1667. doi: 10.1016/0002-9378(94)90418-9. [DOI] [PubMed] [Google Scholar]
- 66.Fried M, Muga RO, Misore AO, Duffy PE. Malaria elicits type 1 cytokines in the human placenta: IFN-gamma and TNF-alpha associated with pregnancy outcomes. JImmunol. 1998;160(5):2523–2530. [PubMed] [Google Scholar]
- 67.Chan PK, Chang AR, Tam WH, Cheung JL, Cheng AF. Prevalence and genotype distribution of cervical human papillomavirus infection: Comparison between pregnant women and non-pregnant controls. J Med Virol. 2002;67:583–588. doi: 10.1002/jmv.10142. [DOI] [PubMed] [Google Scholar]
- 68.Fried M, Duffy PE. Adherence of Plasmodium falciparum to chondroitin sulfate A in the human placenta. Science. 1996;272:1502–1504. doi: 10.1126/science.272.5267.1502. [DOI] [PubMed] [Google Scholar]
- 69.Orozco S, Schmid MA, Parameswaran P, Lachica R, Henn MR, Beatty R, Harris E. Characterization of a model of lethal dengue virus 2 infection in C57BL/6 mice deficient in the alpha/beta interferon receptor. J Gen Virol. 2012;93:2152–2157. doi: 10.1099/vir.0.045088-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Powers C, Fruh K. Rhesus CMV: an emerging animal model for human CMV. Med Microbiol Immunol. 2008;197:109–115. doi: 10.1007/s00430-007-0073-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Golos TG, Bondarenko GI, Dambaeva SV, Breburda EE, Durning M. On the role of placental Major Histocompatibility Complex and decidual leukocytes in implantation and pregnancy success using non-human primate models. Int J Dev Biol. 2010;54:431–443. doi: 10.1387/ijdb.082797tg. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Dambaeva SV, Breburda EE, Durning M, Garthwaite MA, Golos TG. Characterization of decidual leukocyte populations in cynomolgus and vervet monkeys. Journal of Reproductive Immunology. 2009;80:57–69. doi: 10.1016/j.jri.2008.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Slukvin II, Watkins DI, Golos TG. Phenotypic and functional characterization of rhesus monkey decidual lymphocytes: rhesus decidual large granular lymphocytes express CD56 and have cytolytic activity. Journal of Reproductive Immunology. 2001;50:57–79. doi: 10.1016/s0165-0378(00)00090-5. [DOI] [PubMed] [Google Scholar]
- 74.Dambaeva SV, Durning M, Rozner AE, Golos TG. Immunophenotype and cytokine profiles of rhesus monkey CD56bright and CD56dim decidual natural killer cells. Biol Reprod. 2012;86:1–10. doi: 10.1095/biolreprod.111.094383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Novy MJ, Duffy L, Axthelm MK, Sadowsky DW, Witkin SS, Gravett MG, Cassell GH, Waites KB. Ureaplasma parvum or Mycoplasma hominis as sole pathogens cause chorioamnionitis, preterm delivery, and fetal pneumonia in rhesus macaques. Reproductive Sciences. 2009;16:56–70. doi: 10.1177/1933719108325508. [DOI] [PubMed] [Google Scholar]
- 76.Piotrowski P, Croy BA. Maternal cells are widely distributed in murine fetuses in utero. BiolReproduction. 1996;54:1103–1110. doi: 10.1095/biolreprod54.5.1103. [DOI] [PubMed] [Google Scholar]
- 77.Bonney EA, Matzinger P. The maternal immune system's interaction with circulating fetal cells. Journal of Immunology. 1997;158:40–47. [PubMed] [Google Scholar]
- 78.Khosrotehrani K, Johnson KL, Guégan S, Stroh H, Bianchi DW. Natural history of fetal cell microchimerism during and following murine pregnancy. Journal of Reproductive Immunology. 2005;66:1–12. doi: 10.1016/j.jri.2005.02.001. [DOI] [PubMed] [Google Scholar]
- 79.Guimond MJ, Wang B, Fujita J, Terhorst C, Croy BA. Pregnancy-associated uterine granulated metrial gland cells in mutant and transgenic mice. Am J Reprod Immunol. 1996;35:501–509. doi: 10.1111/j.1600-0897.1996.tb00049.x. [DOI] [PubMed] [Google Scholar]
- 80.Lin Y, Zhong Y, Shen W, Chen Y, Shi J, Di J, Zeng S, Saito S. TSLP-induced placental DC activation and IL-10(+) NK cell expansion: comparative study based on BALB/c × C57BL/6 and NOD/SCID × C57BL/6 pregnant models. Clinical Immunology. 2008;126:104–117. doi: 10.1016/j.clim.2007.09.006. [DOI] [PubMed] [Google Scholar]
- 81.Croy BA. Granulated metrial gland cells: hypotheses concerning possible functions during murine gestation. Journal of Reproductive Immunology. 1994;27:85–94. doi: 10.1016/0165-0378(94)90025-6. [DOI] [PubMed] [Google Scholar]
- 82.Madeja Z, Yadi H, Apps R, Boulenouar S, Roper SJ, Gardner L, Moffett A, Colucci F, Hemberger M. Paternal MHC expression on mouse trophoblast affects uterine vascularization and fetal growth. Proc Natl Acad Sci U S A. 2011;108:4012–4017. doi: 10.1073/pnas.1005342108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hiby SE, Apps R, Sharkey AM, Farrell LE, Gardner L, Mulder A, Claas FH, Walker JJ, Redman CW, Morgan L, Tower C, Regan L, Moore GE, Carrington M, Moffett A. Maternal activating KIRs protect against human reproductive failure mediated by fetal HLA-C2. J Clin Invest. 2010;120:4102–4110. doi: 10.1172/JCI43998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Elliott JM, Yokoyama WM. Unifying concepts of MHC-dependent natural killer cell education. Trends Immunol. 2011;32:364–372. doi: 10.1016/j.it.2011.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Murphy SP, Hanna NN, Fast LD, Shaw SK, Berg G, Padbury JF, Romero R, Sharma S. Evidence for participation of uterine natural killer cells in the mechanisms responsible for spontaneous preterm labor and delivery. American Journal of Obstetrics & Gynecology. 2009;200:308–309. doi: 10.1016/j.ajog.2008.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kahn DA, Baltimore D. Pregnancy induces a fetal antigen-specific maternal T regulatory cell response that contributes to tolerance. Proceedings of the National Academy of Sciences. 2010;107:9299–9304. doi: 10.1073/pnas.1003909107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature. 2012;490:102–106. doi: 10.1038/nature11462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bonney EA, Shepard MT, Bizargity P. Transient modification within a pool of CD4 T cells in the maternal spleen. Immunology. 2011;134:270–280. doi: 10.1111/j.1365-2567.2011.03486.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Norton MT, Fortner KA, Oppenheimer KH, Bonney EA. Evidence that CD8 T-cell homeostasis and function remain intact during murine pregnancy. Immunology. 2010;131:426–437. doi: 10.1111/j.1365-2567.2010.03316.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Moldenhauer LM, Hayball JD, Robertson SA. Utilising T cell receptor transgenic mice to define mechanisms of maternal T cell tolerance in pregnancy. Journal of Reproductive Immunology. 2010;87:1–13. doi: 10.1016/j.jri.2010.05.007. [DOI] [PubMed] [Google Scholar]
- 91.Bizargity P, Bonney EA. Dendritic cells: a family portrait at mid-gestation. Immunology. 2009;126:565–578. doi: 10.1111/j.1365-2567.2008.02918.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Erlebacher A, Vencato D, Price KA, Zhang D, Glimcher LH. Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. Journal of Clinical Investigation. 2007;117:1399–1411. doi: 10.1172/JCI28214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Padilla-Carlin DJ, McMurray DN, Hickey AJ. The guinea pig as a model of infectious diseases. Comp Med. 2008;58:324–340. [PMC free article] [PubMed] [Google Scholar]
- 94.Hu X, Wu GP, Zhang MH, Pan SQ, Wang RR, Ouyang JH, Liu JG, Chen ZY, Tian H, Liu DB. GC-MS-based metabolic profiling reveals metabolic changes in anaphylaxis animal models. Anal Bioanal Chem. 2012;404:887–893. doi: 10.1007/s00216-012-6129-x. [DOI] [PubMed] [Google Scholar]
- 95.Schäfer H, Burger R. Tools for cellular immunology and vaccine research the in the guinea pig: Monoclonal antibodies to cell surface antigens and cell lines. Vaccine. 2012;30:5804–5811. doi: 10.1016/j.vaccine.2012.07.012. [DOI] [PubMed] [Google Scholar]
- 96.Gertz EM, Agarwala R, Mage RG, Schäffer AA. Comparative Analysis of Genome Sequences of the Th2 Cytokine Region of Rabbit (Oryctolagus cuniculus) with those of Nine Different Species. Immunology and Immunogenetics Insights. 2011;3:59–82. doi: 10.4137/III.S7236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wattegedera S, Rocchi M, Sales J, Howard CJ, Hope JC, Entrican G. Antigen-specific peripheral immune responses are unaltered during normal pregnancy in sheep. Journal of Reproductive Immunology. 2008;77:171–178. doi: 10.1016/j.jri.2007.07.003. [DOI] [PubMed] [Google Scholar]
- 98.Assal-Meliani A, Charpigny G, Reinaud P, Martal J, Chaouat G. Recombinant ovine trophoblastin (roTP) inhibits ovine, murine and human lymphocyte proliferation. Journal of Reproductive Immunology. 1993;25:149–165. doi: 10.1016/0165-0378(93)90055-m. [DOI] [PubMed] [Google Scholar]
- 99.Mincheva-Nilsson L. Pregnancy and gamma/delta T cells: taking on the hard questions. Reprod Biol Endocrinol. 2003;1:120. doi: 10.1186/1477-7827-1-120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Fan DX, Duan J, Li MQ, Xu B, Li DJ, Jin LP. The decidual gamma-delta T cells up-regulate the biological functions of trophoblasts via IL-10 secretion in early human pregnancy. Clin Immunol. 2011;141:284–292. doi: 10.1016/j.clim.2011.07.008. [DOI] [PubMed] [Google Scholar]
- 101.Exley MA, Boyson JE. Protective role of regulatory decidual gammadelta T cells in pregnancy. Clin Immunol. 2011;141:236–239. doi: 10.1016/j.clim.2011.09.004. [DOI] [PubMed] [Google Scholar]
- 102.Fox A, Maddox JF, de Veer MJ, Meeusen EN. GammadeltaTCR+ cells of the pregnant ovine uterus express variable T cell receptors and contain granulysin. Journal of Reproductive Immunology. 2010;84:52–56. doi: 10.1016/j.jri.2009.10.003. [DOI] [PubMed] [Google Scholar]
- 103.Majewski AC, Tekin S, Hansen PJ. Local versus systemic control of numbers of endometrial T cells during pregnancy in sheep. Immunology. 2001;102:317–322. doi: 10.1046/j.1365-2567.2001.01182.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Engelhardt H, Croy BA, King GJ. Role of uterine immune cells in early pregnancy in pigs. J Reprod Fertil Suppl. 1997;52:115–131. [PubMed] [Google Scholar]
- 105.Clancy B, Finlay BL, Darlington RB, Anand KJ. Extrapolating brain development from experimental species to humans. NeuroToxicology. 2007;28:931–937. doi: 10.1016/j.neuro.2007.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zhou Q, Renard JP, Le Friec G, Brochard V, Beaujean N, Cherifi Y, Fraichard A, Cozzi J. Generation of fertile cloned rats by regulating oocyte activation. Science. 2003;302:1179. doi: 10.1126/science.1088313. [DOI] [PubMed] [Google Scholar]
- 107.Dickinson H, Walker DW, Cullen-McEwen L, Wintour EM, Moritz K. The spiny mouse (Acomys cahirinus) completes nephrogenesis before birth. Am J Physiol Renal Physiol. 2005;289:F273–F279. doi: 10.1152/ajprenal.00400.2004. [DOI] [PubMed] [Google Scholar]
- 108.Seifert AW, Kiama SG, Seifert MG, Goheen JR, Palmer TM, Maden M. Skin shedding and tissue regeneration in African spiny mice (Acomys) Nature. 2012;489:561–565. doi: 10.1038/nature11499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Anthony RV, Scheaffer AN, Wright CD, Regnault TR. Ruminant models of prenatal growth restriction. Reprod Suppl. 2003;61:183–194. [PubMed] [Google Scholar]
- 110.Sibai B, Dekker G, Kupferminc M. Pre-eclampsia Primary, secondary, and tertiary prevention of pre-eclampsia. Lancet. 2005;365:785–799. doi: 10.1016/S0140-6736(05)17987-2. [DOI] [PubMed] [Google Scholar]
- 111.Jacobs M, Nassar N, Roberts CL, Hadfield R, Morris JM, Ashton AW. Levels of soluble fms-like tyrosine kinase one in first trimester and outcomes of pregnancy: a systematic review. Reprod Biol Endocrinol. 2011;9:77. doi: 10.1186/1477-7827-9-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Brosens IA, Robertson WB, Dixon HG. The role of the spiral arteries in the pathogenesis of preeclampsia. Obstetrics & Gynecology Annual. 1972;1:177–191. [PubMed] [Google Scholar]
- 113.Bernstein IM, Meyer MC, Osol G, Ward K. Intolerance to volume expansion: a theorized mechanism for the development of preeclampsia. Obstet Gynecol. 1998;92:306–308. doi: 10.1016/s0029-7844(98)00207-5. [DOI] [PubMed] [Google Scholar]
- 114.Raynor BD, Bonney EA, Jang KT, Coto W, Garcia MS. Preeclampsia and Chlamydia pneumoniae: is there a link? Hypertension in Pregnancy. 2004;23:129–134. doi: 10.1081/PRG-120028284. [DOI] [PubMed] [Google Scholar]
- 115.Kim YM, Romero R, Oh SY, Kim CJ, Kilburn BA, Armant DR, Nien JK, Gomez R, Mazor M, Saito S, Abrahams VM, Mor G. Toll-like receptor 4: a potential link between "danger signals," the innate immune system, and preeclampsia? American Journal of Obstetrics & Gynecology. 2005;193:921–927. doi: 10.1016/j.ajog.2005.07.076. [DOI] [PubMed] [Google Scholar]
- 116.Bonney EA. Preeclampsia: a view through the danger model. Journal of Reproductive Immunology. 2007;76:68–74. doi: 10.1016/j.jri.2007.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Redman CW, Sargent IL. Immunology of pre-eclampsia. Am J Reprod Immunol. 2010;63:534–543. doi: 10.1111/j.1600-0897.2010.00831.x. [DOI] [PubMed] [Google Scholar]
- 118.Roberts JM, Taylor RN, Goldfien A. Clinical and biochemical evidence of endothelial cell dysfunction in the pregnancy syndrome preeclampsia. Am J Hypertens. 1991;4:700–708. doi: 10.1093/ajh/4.8.700. [DOI] [PubMed] [Google Scholar]
- 119.Ilekis JV, Reddy UM, Roberts JM. Preeclampsia--a pressing problem: an executive summary of a National Institute of Child Health and Human Development workshop. Reprod Sci. 2007;14:508–523. doi: 10.1177/1933719107306232. [DOI] [PubMed] [Google Scholar]
- 120.McCarthy FP, Kingdom JC, Kenny LC, Walsh SK. Animal models of preeclampsia; uses and limitations. Placenta. 2011;32:413–419. doi: 10.1016/j.placenta.2011.03.010. [DOI] [PubMed] [Google Scholar]
- 121.Stout C, Lemmon WB. Glomerular capillary endothelial swelling in a pregnant chimpanzee. Am J Obstet Gynecol. 1969;105:212–215. doi: 10.1016/0002-9378(69)90060-x. [DOI] [PubMed] [Google Scholar]
- 122.Sunderland NS, Thomson SE, Heffernan SJ, Lim S, Thompson J, Ogle R, McKenzie P, Kirwan PJ, Makris A, Hennessy A. Tumor necrosis factor alpha induces a model of preeclampsia in pregnant baboons (Papio hamadryas) Cytokine. 2011;56:192–199. doi: 10.1016/j.cyto.2011.06.003. [DOI] [PubMed] [Google Scholar]
- 123.Orange S, Rasko JE, Thompson JF, Vaughan J, Olive E, Pedler M, Horvath JS, Hennessy A. Interleukin-10 regulates arterial pressure in early primate pregnancy. Cytokine. 2005;29:176–185. doi: 10.1016/j.cyto.2004.10.011. [DOI] [PubMed] [Google Scholar]
- 124.Davisson RL, Hoffmann DS, Butz GM, Aldape G, Schlager G, Merrill DC, Sethi S, Weiss RM, Bates JN. Discovery of a Spontaneous Genetic Mouse Model of Preeclampsia. Hypertension. 2002;39:337–342. doi: 10.1161/hy02t2.102904. [DOI] [PubMed] [Google Scholar]
- 125.Dokras A, Hoffmann DS, Eastvold JS, Kienzle MF, Gruman LM, Kirby PA, Weiss RM, Davisson RL. Severe Feto-Placental Abnormalities Precede the Onset of Hypertension and Proteinuria in a Mouse Model of Preeclampsia. Biology of Reproduction. 2006;75:899–907. doi: 10.1095/biolreprod.106.053603. [DOI] [PubMed] [Google Scholar]
- 126.Sharkey LC, Kirchain S, McCune SA, Simpson GI, Archambault EZ, Boatright NK, Hicks E, Fray J. Progesterone increases blood pressure in spontaneous gestational hypertension in rats. Am J Hypertens. 2005;18:36–43. doi: 10.1016/j.amjhyper.2004.07.024. [DOI] [PubMed] [Google Scholar]
- 127.Furuya M, Ishida J, Inaba S, Kasuya Y, Kimura S, Nemori R, Fukamizu A. Impaired placental neovascularization in mice with pregnancy-associated hypertension. Lab Invest. 2008;88:416–429. doi: 10.1038/labinvest.2008.7. [DOI] [PubMed] [Google Scholar]
- 128.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. doi: 10.1016/j.placenta.2010.01.011. [DOI] [PubMed] [Google Scholar]
- 129.Bohlender J, Ganten D, Luft FC. Rats transgenic for human renin and human angiotensinogen as a model for gestational hypertension. Journal of the American Society of Nephrology. 2056;11:2056–2061. doi: 10.1681/ASN.V11112056. [DOI] [PubMed] [Google Scholar]
- 130.Young J. The AEtiology of Eclampsia and Albuminuria and their Relation to Accidental Haemorrhage: (An Anatomical and Experimental Investigation.) Proc R Soc Med. 1914;7:307–348. [PMC free article] [PubMed] [Google Scholar]
- 131.Li J, LaMarca B, Reckelhoff JF. A model of preeclampsia in rats: the reduced uterine perfusion pressure (RUPP) model. Am J Physiol Heart Circ Physiol. 2012;303:H1–H8. doi: 10.1152/ajpheart.00117.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Douglas BH. The rat as a model for preeclampsia. Perspect Nephrol Hypertens. 1976;5:411–419. [PubMed] [Google Scholar]
- 133.Ianosi-Irimie M, Vu HV, Whitbred JM, Pridjian CA, Nadig JD, Williams MY, Wrenn DC, Pridjian G, Puschett JB. A rat model of preeclampsia. Clin Exp Hypertens. 2005;27:605–617. doi: 10.1080/10641960500298608. [DOI] [PubMed] [Google Scholar]
- 134.Yallampalli C, Garfield RE. Inhibition of nitric oxide synthesis in rats during pregnancy produces signs similar to those of preeclampsia. Am J Obstet Gynecol. 1993;169:1316–1320. doi: 10.1016/0002-9378(93)90299-x. [DOI] [PubMed] [Google Scholar]
- 135.Maynard SE, Min J-Y, Merchan J, Lim K-H, Li J, Mondal S, Libermann TA, Morgan JP, Sellke FW, Stillman IE, Epstein FH, Sukhatme VP, Karumanchi SA. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. The Journal of Clinical Investigation. 2003;111:649–658. doi: 10.1172/JCI17189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, Bdolah Y, Lim KH, Yuan HT, Libermann TA, Stillman IE, Roberts D, D'Amore PA, Epstein FH, Sellke FW, Romero R, Sukhatme VP, Letarte M, Karumanchi SA. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med. 2006;12:642–649. doi: 10.1038/nm1429. [DOI] [PubMed] [Google Scholar]
- 137.Seidl DC, Hughes HC, Bertolet R, Lang CM. True pregnancy toxemia (preeclampsia) in the guinea pig (Cavia porcellus) Lab Anim Sci. 1979;29:472–478. [PubMed] [Google Scholar]
- 138.Golden JG, Hughes HC, Lang CM. Experimental toxemia in the pregnant guinea pig (Cavia porcellus) Lab Anim Sci. 1980;30:174–179. [PubMed] [Google Scholar]
- 139.Abitbol MM, Gallo GR, Pirani CL, Ober WB. Production of experimental toxemia in the pregnant rabbit. Am J Obstet Gynecol. 1976;124:460–470. doi: 10.1016/0002-9378(76)90169-1. [DOI] [PubMed] [Google Scholar]
- 140.Al-Qudah KM. Oxidant and antioxidant profile of hyperketonemic ewes affected by pregnancy toxemia. Vet Clin Pathol. 2011;40:60–65. doi: 10.1111/j.1939-165X.2011.00284.x. [DOI] [PubMed] [Google Scholar]
- 141.Yarim GF, Ciftci G. Serum protein pattern in ewe with pregnancy toxemia. Vet Res Commun. 2009;33:431–438. doi: 10.1007/s11259-008-9189-9. [DOI] [PubMed] [Google Scholar]
- 142.Bukowski R. Stillbirth and fetal growth restriction. [Review] Clinical Obstetrics & Gynecology. 2010;53:673–680. doi: 10.1097/GRF.0b013e3181eba0db. [DOI] [PubMed] [Google Scholar]
- 143.Wu G, Bazer FW, Wallace JM, Spencer TE. BOARD-INVITED REVIEW: Intrauterine growth retardation: Implications for the animal sciences. J Anim Sci. 2006;84:2316–2337. doi: 10.2527/jas.2006-156. [DOI] [PubMed] [Google Scholar]
- 144.McDonald TJ, Wu G, Nijland MJ, Jenkins SL, Nathanielsz PW, Jansson T. Effect of 30 % nutrient restriction in the first half of gestation on maternal and fetal baboon serum amino acid concentrations. Br J Nutr. 2012;9:1–7. doi: 10.1017/S0007114512003261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Roberts VHJ, Räsänen JP, Novy MJ, Frias A, Louey S, Morgan TK, Thornburg KL, Spindel ER, Grigsby PL. Restriction of placental vasculature in a non-human primate: A unique model to study placental plasticity. Placenta. 2012;33:73–76. doi: 10.1016/j.placenta.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Royston JP, Flecknell PA, Wootton R. New evidence that the intra-uterine growth-retarded piglet is a member of a discrete subpopulation. Biol Neonate. 1982;42:100–104. doi: 10.1159/000241582. [DOI] [PubMed] [Google Scholar]
- 147.Ruster M, Sommer M, Stein G, Bauer K, Walter B, Wolf G, Bauer R. Renal Angiotensin receptor type 1 and 2 upregulation in intrauterine growth restriction of newborn piglets. Cells Tissues Organs. 2006;182:106–114. doi: 10.1159/000093065. [DOI] [PubMed] [Google Scholar]
- 148.Hefler LA, Reyes CA, O'Brien WE, Gregg AR. Perinatal development of endothelial nitric oxide synthase-deficient mice. Biol Reprod. 2001;64:666–673. doi: 10.1095/biolreprod64.2.666. [DOI] [PubMed] [Google Scholar]
- 149.Hefler LA, Tempfer CB, Moreno RM, O'Brien WE, Gregg AR. Endothelial-derived nitric oxide and angiotensinogen: blood pressure and metabolism during mouse pregnancy. Am J Physiol Regul Integr Comp Physiol. 2001;280:R174–R182. doi: 10.1152/ajpregu.2001.280.1.R174. [DOI] [PubMed] [Google Scholar]
- 150.Puglianiello A, Germani D, Antignani S, Tomba GS, Cianfarani S. Changes in the expression of hypothalamic lipid sensing genes in rat model of intrauterine growth retardation (IUGR) Pediatric Research. 2007;61:433–437. doi: 10.1203/pdr.0b013e3180332d4e. [DOI] [PubMed] [Google Scholar]
- 151.Turner AJ, Trudinger BJ. A Modification of the Uterine Artery Restriction Technique in the Guinea Pig Fetus Produces Asymmetrical Ultrasound Growth. Placenta. 2009;30:236–240. doi: 10.1016/j.placenta.2008.11.023. [DOI] [PubMed] [Google Scholar]
- 152.Eixarch E, Batalle D, Illa M, Munoz-Moreno E, Arbat-Plana A, Amat-Roldan I, Figueras F, Gratacos E. Neonatal neurobehavior and diffusion MRI changes in brain reorganization due to intrauterine growth restriction in a rabbit model. PLoS ONE. 2012;7:e31497. doi: 10.1371/journal.pone.0031497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Lecarpentier E, Morel O, Tarrade A, Dahirel M, Bonneau M, Gayat E, Evain-Brion D, Chavatte-Palmer P, Tsatsaris V. Quantification of utero-placental vascularization in a rabbit model of IUGR with three-dimensional power Doppler angiography. Placenta. 2012;33:769–775. doi: 10.1016/j.placenta.2012.06.013. [DOI] [PubMed] [Google Scholar]
- 154.Sebert SP, Dellschaft NS, Chan LLY, Street H, Henry M, Francois C, Sharma V, Fainberg HP, Patel N, Roda J, Keisler D, Budge H, Symonds ME. Maternal Nutrient Restriction During Late Gestation and Early Postnatal Growth in Sheep Differentially Reset the Control of Energy Metabolism in the Gastric Mucosa. Endocrinology. 2011;152:2816–2826. doi: 10.1210/en.2011-0169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Laurent A, Pelage J-P, Wassef M, Martal J. Fertility after bilateral uterine artery embolization in a sheep model. Fertil Steril. 2008;89:1371–1383. doi: 10.1016/j.fertnstert.2007.03.058. [DOI] [PubMed] [Google Scholar]
- 156.Robinson JS, Kingston EJ, Jones CT, Thorburn GD. Studies on experimental growth retardation in sheep. The effect of removal of a endometrial caruncles on fetal size and metabolism. J Dev Physiol. 1979;1:379–398. [PubMed] [Google Scholar]
- 157.Arroyo JA, Anthony RV, Parker TA, Galan HL. Differential expression of placental and vascular endothelial nitric oxide synthase in an ovine model of fetal growth restriction. American Journal of Obstetrics & Gynecology. 2006;195:771–777. doi: 10.1016/j.ajog.2006.06.018. [DOI] [PubMed] [Google Scholar]
- 158.Vuguin PM. Animal models for small for gestational age and fetal programming of adult disease. Horm Res. 2007;68:113–123. doi: 10.1159/000100545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Ziebell BT, Galan HL, Anthony RV, Regnault TRH, Parker TA, Arroyo JA. Ontogeny of endothelial nitric oxide synthase mRNA in an ovine model of fetal and placental growth restriction. American Journal of Obstetrics and Gynecology. 2007;197:420.e421–420.e425. doi: 10.1016/j.ajog.2007.07.016. [DOI] [PubMed] [Google Scholar]
- 160.Galan HL, Regnault TR, Le Cras TD, Tyson RW, Anthony RV, Wilkening RB, Abman SH. Cotyledon and binucleate cell nitric oxide synthase expression in an ovine model of fetal growth restriction. J Appl Physiol. 2001;90:2420–2426. doi: 10.1152/jappl.2001.90.6.2420. [DOI] [PubMed] [Google Scholar]
- 161.Robinson JS, Jones CT, Kingston EJ. Studies on experimental growth retardation in sheep. The effects of maternal hypoxaemia. J Dev Physiol. 1983;5:89–100. [PubMed] [Google Scholar]
- 162.Cummings AM, Kavlock RJ. Gene-environment interactions: a review of effects on reproduction and development. Crit Rev Toxicol. 2004;34:461–485. doi: 10.1080/10408440490519786. [DOI] [PubMed] [Google Scholar]
- 163.Craig M. Stress and recurrent miscarriage. Stress. 2001;4:205–213. doi: 10.3109/10253890109035019. [DOI] [PubMed] [Google Scholar]
- 164.Mathias S, Allen WR. Immunization with stallion lymphocytes for treatment of recurrent spontaneous abortion in thoroughbred mares. J Reprod Fertil Suppl. 2000;56:645–650. [PubMed] [Google Scholar]
- 165.Croy BA, Wessels JM, Linton NF, van den Heuvel M, Edwards AK, Tayade C. Cellular and molecular events in early and mid gestation porcine implantation sites: a review. Soc Reprod Fertil Suppl. 2009;66:233–244. [PubMed] [Google Scholar]
- 166.Nakashima A, Shima T, Inada K, Ito M, Saito S. The balance of the immune system between T cells and NK cells in miscarriage. Am J Reprod Immunol. 2012;67:304–310. doi: 10.1111/j.1600-0897.2012.01115.x. [DOI] [PubMed] [Google Scholar]
- 167.Nielsen HS. Secondary recurrent miscarriage and H-Y immunity. Hum Reprod Update. 2011;17:558–574. doi: 10.1093/humupd/dmr005. [DOI] [PubMed] [Google Scholar]
- 168.Bansal AS, Bajardeen B, Thum MY. The basis and value of currently used immunomodulatory therapies in recurrent miscarriage. Journal of Reproductive Immunology. 2012;93:41–51. doi: 10.1016/j.jri.2011.10.002. [DOI] [PubMed] [Google Scholar]
- 169.Knapp LA, Ha JC, Sackett GP. Parental MHC antigen sharing and pregnancy wastage in captive pigtailed macaques. Journal of Reproductive Immunology. 1996;32:73–88. doi: 10.1016/s0165-0378(96)00988-6. [DOI] [PubMed] [Google Scholar]
- 170.Owiti GE, Tarantal AF, Lasley BL, Hendrickx AG. The effect of the anti-progestin RU 486 on early pregnancy in the long-tailed macaque (Macaca fascicularis) Contraception. 1989;40:201–211. doi: 10.1016/0010-7824(89)90007-3. [DOI] [PubMed] [Google Scholar]
- 171.Smith MA, Takeuchi K, Brackett RE, McClure HM, Raybourne RB, Williams KM, Babu US, Ware GO, Broderson JR, Doyle MP. Nonhuman primate model for Listeria monocytogenes-induced stillbirths. Infect Immun. 2003;71:1574–1579. doi: 10.1128/IAI.71.3.1574-1579.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Ho HN, Chen SU, Yang YS, Huang SC, Lee TY, Gill TJ., 3rd Age, environment, and lymphocyte immunization influence the spontaneous resorption rate in the CBA/J × DBA/2J mouse model. Am J Reprod Immunol. 1994;31:47–51. doi: 10.1111/j.1600-0897.1994.tb00846.x. [DOI] [PubMed] [Google Scholar]
- 173.Haddad EK, Duclos AJ, Baines MG. Early embryo loss is associated with local production of nitric oxide by decidual mononuclear cells. JExpMed. 1995;182:1143–1152. doi: 10.1084/jem.182.4.1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Clark DA, Chaput A, Tutton D. Active suppression of host-vs-graft reaction in pregnant mice. VII. Spontaneous abortion of allogeneic CBA/J × DBA/2 fetuses in the uterus of CBA/J mice correlates with deficient non-T suppressor cell activity. The Journal of Immunology. 1986;136:1668–1675. [PubMed] [Google Scholar]
- 175.Arck PC, Merali FS, Manuel J, Chaouat G, Clark DA. Stress-triggered abortion: inhibition of protective suppression and promotion of tumor necrosis factor-alpha (TNF-alpha) release as a mechanism triggering resorptions in mice. Am J Reprod Immunol. 1995;33:74–80. doi: 10.1111/j.1600-0897.1995.tb01141.x. [DOI] [PubMed] [Google Scholar]
- 176.Toder V, Strassburger D, Carp H, Irlin I. Mouse model for the treatment of immune pregnancy loss. Am J Reprod Immunol. 1991;26:42–46. doi: 10.1111/j.1600-0897.1991.tb00701.x. [DOI] [PubMed] [Google Scholar]
- 177.Clark DA, Manuel J, Lee L, Chaouat G, Gorczynski RM, Levy GA. Ecology of danger-dependent cytokine-boosted spontaneous abortion in the CBA × DBA/2 mouse model. I. Synergistic effect of LPS and (TNF-alpha + IFN-gamma) on pregnancy loss. Am J Reprod Immunol. 2004;52:370–378. doi: 10.1111/j.1600-0897.2004.00237.x. [DOI] [PubMed] [Google Scholar]
- 178.Chaouat G, Menu E, Delage G, Moreau JF, Khrishnan L, Hui L, Meliani AA, Martal J, Raghupathy R, Lelaidier C, et al. Immuno-endocrine interactions in early pregnancy. Hum Reprod. 1995;10(Suppl 2):55–59. doi: 10.1093/humrep/10.suppl_2.55. [DOI] [PubMed] [Google Scholar]
- 179.Chaouat G, Meliani AA, Martal J, Raghupathy R, Eliot J, Mosmann T, Wegmann TG. IL-10 prevents naturally occuring fetal loss in the CBAxDBA/2 mating combination, and local defect in IL-10 production in the abortion-prone combination is corrected by in vivo injection of IFN-τ. JImmunol. 1995;154:4261–4268. [PubMed] [Google Scholar]
- 180.Zenclussen AC, Gerlof K, Zenclussen ML, Ritschel S, Zambon Bertoja A, Fest S, Hontsu S, Ueha S, Matsushima K, Leber J, Volk H-D. Regulatory T cells induce a privileged tolerant microenvironment at the fetal-maternal interface. [see comment] European Journal of Immunology. 2006;36:82–94. doi: 10.1002/eji.200535428. [DOI] [PubMed] [Google Scholar]
- 181.Wafula PO, Teles A, Schumacher A, Pohl K, Yagita H, Volk HD, Zenclussen AC. PD-1 but not CTLA-4 blockage abrogates the protective effect of regulatory T cells in a pregnancy murine model. Am J Reprod Immunol. 2009;62:283–292. doi: 10.1111/j.1600-0897.2009.00737.x. [DOI] [PubMed] [Google Scholar]
- 182.Dixon ME, Chien EK, Osol G, Callas PW, Bonney EA. Failure of decidual arteriolar remodeling in the CBA/J × DBA/2 murine model of recurrent pregnancy loss is linked to increased expression of tissue inhibitor of metalloproteinase 2 (TIMP-2) American Journal of Obstetrics & Gynecology. 2006;194:113–119. doi: 10.1016/j.ajog.2005.06.063. [DOI] [PubMed] [Google Scholar]
- 183.Redecha P, van Rooijen N, Torry D, Girardi G. Pravastatin prevents miscarriages in mice: role of tissue factor in placental and fetal injury. Blood. 2009;113:4101–4109. doi: 10.1182/blood-2008-12-194258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Chaouat G, Kiger N, Wegmann TG. Vaccination against spontaneous abortion in mice. Journal of Reproductive Immunology. 1983;5:389–392. doi: 10.1016/0165-0378(83)90248-6. [DOI] [PubMed] [Google Scholar]
- 185.Jin LP, Zhou YH, Zhu XY, Wang MY, Li DJ. Adoptive transfer of paternal antigen-hyporesponsive T cells facilitates a Th2 bias in peripheral lymphocytes and at materno-fetal interface in murine abortion-prone matings. Am J Reprod Immunol. 2006;56:258–266. doi: 10.1111/j.1600-0897.2006.00425.x. [DOI] [PubMed] [Google Scholar]
- 186.Bertoja AZ, Zenclussen ML, Casalis PA, Sollwedel A, Schumacher A, Woiciechowsky C, Volk HD, Zenclussen AC. Anti-P- and E-selectin therapy prevents abortion in the CBA/J × DBA/2J combination by blocking the migration of Th1 lymphocytes into the foetal-maternal interface. Cellular Immunology. 2005;238:97–102. doi: 10.1016/j.cellimm.2006.02.002. [DOI] [PubMed] [Google Scholar]
- 187.Zhao F-X, Zhang Y-Y, Liu R-H, Li S-M. Effect of blockage of costimulatory signal on murine abortion-prone model. Chinese Medical Journal. 2007;120:1247–1250. [PubMed] [Google Scholar]
- 188.Samstein Robert M, Josefowicz Steven Z, Arvey A, Treuting Piper M, Rudensky Alexander Y. Extrathymic Generation of Regulatory T Cells in Placental Mammals Mitigates Maternal-Fetal Conflict. Cell. 2012;150:29–38. doi: 10.1016/j.cell.2012.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Boyson JE, Nagarkatti N, Nizam L, Exley MA, Strominger JL. Gestation stage-dependent mechanisms of invariant natural killer T cell-mediated pregnancy loss. Proc Natl Acad Sci U S A. 2006;103:4580–4585. doi: 10.1073/pnas.0511025103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Erlebacher A, Zhang D, Parlow AF, Glimcher LH. Ovarian insufficiency and early pregnancy loss induced by activation of the innate immune system. Journal of Clinical Investigation. 2004;114:39–48. doi: 10.1172/JCI20645. [see comment]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Tayade C, Fang Y, Croy BA. A review of gene expression in porcine endometrial lymphocytes, endothelium and trophoblast during pregnancy success and failure. J Reprod Dev. 2007;53:455–463. doi: 10.1262/jrd.18170. [DOI] [PubMed] [Google Scholar]
- 192.Harrison CJ, Caruso N. Correlation of maternal and pup NK-like activity and TNF responses against cytomegalovirus to pregnancy outcome in inbred guinea pigs. Journal of Medical Virology. 2000;60:230–236. [PubMed] [Google Scholar]
- 193.Rodolakis A, Salinas J, Papp J. Recent advances on ovine chlamydial abortion. Vet Res. 1998;29:275–288. [PubMed] [Google Scholar]
- 194.Pierangeli SS, Harris EN. Induction of phospholipid-binding antibodies in mice and rabbits by immunization with human beta 2 glycoprotein 1 or anticardiolipin antibodies alone. Clin Exp Immunol. 1993;93:269–272. doi: 10.1111/j.1365-2249.1993.tb07978.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Elovitz MA. Animal models of pre-term birth. Havemeyer Foundation Monograph Series. 2008 [Google Scholar]
- 196.Kamath-Rayne BD, Defranco EA, Chung E, Chen A. Subtypes of Preterm Birth and the Risk of Postneonatal Death. J Pediatr. 2012;7:7. doi: 10.1016/j.jpeds.2012.06.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Mercer BM, Macpherson CA, Goldenberg RL, Goepfert AR, Hauguel-de Mouzon S, Varner MW, Iams JD, Meis PJ, Moawad AH, Miodovnik M, Caritis SN, Van Dorsten JP, Sorokin Y, Thurnau GR, Spong CY. Are women with recurrent spontaneous preterm births different from those without such history? Am J Obstet Gynecol. 2006;194:1176–1184. doi: 10.1016/j.ajog.2006.01.069. discussion 1184-1175. [DOI] [PubMed] [Google Scholar]
- 198.Schuit E, Stock S, Groenwold RH, Maurel K, Combs CA, Garite T, Spong CY, Thom EA, Rouse DJ, Caritis SN, Saade GR, Zachary JM, Norman JE, Rode L, Klein K, Tabor A, Cetingoz E, Morrison JC, Magann EF, Briery CM, Serra V, Perales A, Meseguer J, Nassar AH, Lim AC, Moons KG, Kwee A, Mol BW. Progestogens to prevent preterm birth in twin pregnancies: an individual participant data meta-analysis of randomized trials. BMC Pregnancy Childbirth. 2012;12:13. doi: 10.1186/1471-2393-12-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Gervasi MT, Romero R, Bracalente G, Erez O, Dong Z, Hassan SS, Yeo L, Yoon BH, Chaiworapongsa T. Midtrimester amniotic fluid concentrations of interleukin-6 and interferon-gamma-inducible protein-10: evidence for heterogeneity of intra-amniotic inflammation and associations with spontaneous early (<32 weeks) and late (>32 weeks) preterm delivery. J Perinat Med. 2012;40:329–343. doi: 10.1515/jpm-2012-0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Romero R, Chaiworapongsa T, Alpay Savasan Z, Xu Y, Hussein Y, Dong Z, Kusanovic JP, Kim CJ, Hassan SS. Damage-associated molecular patterns (DAMPs) in preterm labor with intact membranes and preterm PROM: a study of the alarmin HMGB1. J Matern Fetal Neonatal Med. 2011;24:1444–1455. doi: 10.3109/14767058.2011.591460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Fonseca EB, Celik E, Parra M, Singh M, Nicolaides KH. Progesterone and the risk of preterm birth among women with a short cervix. N Engl J Med. 2007;357:462–469. doi: 10.1056/NEJMoa067815. [DOI] [PubMed] [Google Scholar]
- 202.Sanchez SE, Alva AV, Diez Chang G, Qiu C, Yanez D, Gelaye B, Williams MA. Risk of Spontaneous Preterm Birth in Relation to Maternal Exposure to Intimate Partner Violence During Pregnancy in Peru. Matern Child Health J. 2012;17:17. doi: 10.1007/s10995-012-1012-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Aliaga S, Price W, McCaffrey M, Ivester T, Boggess K, Tolleson-Rinehart S. Practice variation in late-preterm deliveries: a physician survey. J Perinatol. 2012;27:119. doi: 10.1038/jp.2012.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Murray SA, Morgan JL, Kane C, Sharma Y, Heffner CS, Lake J, Donahue LR. Mouse gestation length is genetically determined. PLoS ONE. 2010;5:e12418. doi: 10.1371/journal.pone.0012418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Robertson SA, Christiaens I, Dorian CL, Zaragoza DB, Care AS, Banks AM, Olson DM. Interleukin-6 is an essential determinant of on-time parturition in the mouse. Endocrinology. 2010;151:3996–4006. doi: 10.1210/en.2010-0063. [DOI] [PubMed] [Google Scholar]
- 206.Meis PJ, Klebanoff M, Thom E, Dombrowski MP, Sibai B, Moawad AH, Spong CY, Hauth JC, Miodovnik M, Varner MW, Leveno KJ, Caritis SN, Iams JD, Wapner RJ, Conway D, O'Sullivan MJ, Carpenter M, Mercer B, Ramin SM, Thorp JM, Peaceman AM, Gabbe S. Prevention of recurrent preterm delivery by 17 alpha-hydroxyprogesterone caproate. N Engl J Med. 2003;348:2379–2385. doi: 10.1056/NEJMoa035140. [DOI] [PubMed] [Google Scholar]
- 207.Hassan SS, Romero R, Vidyadhari D, Fusey S, Baxter JK, Khandelwal M, Vijayaraghavan J, Trivedi Y, Soma-Pillay P, Sambarey P, Dayal A, Potapov V, O'Brien J, Astakhov V, Yuzko O, Kinzler W, Dattel B, Sehdev H, Mazheika L, Manchulenko D, Gervasi MT, Sullivan L, Conde-Agudelo A, Phillips JA, Creasy GW for the PT. Vaginal progesterone reduces the rate of preterm birth in women with a sonographic short cervix: a multicenter, randomized, double-blind, placebo-controlled trial. Ultrasound in Obstetrics & Gynecology. 2011;38:18–31. doi: 10.1002/uog.9017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Bastek J, Adamczak J, Hoffman S, Elovitz M, Srinivas S. Trends in Prematurity: What do Changes at an Urban Institution Suggest About the Public Health Impact of 17-Alpha Hydroxyprogesterone Caproate? Maternal and Child Health Journal. 2012;16:564–568. doi: 10.1007/s10995-011-0783-z. [DOI] [PubMed] [Google Scholar]
- 209.Manuck TA, Lai Y, Meis PJ, Dombrowski MP, Sibai B, Spong CY, Rouse DJ, Durnwald CP, Caritis SN, Wapner RJ, Mercer BM, Ramin SM. Progesterone receptor polymorphisms and clinical response to 17-alpha-hydroxyprogesterone caproate. Am J Obstet Gynecol. 2011;205:135 e131–135.e139. doi: 10.1016/j.ajog.2011.03.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Gorenberg D, Beharry K, Nishihara KC, Chang E, Waltzman J, Akmal A, Asrat T. Dose response of RU486 in a novel rabbit model of noninfectious preterm birth: comparative efficacy of 3 routes of administration. American Journal of Obstetrics & Gynecology. 2005;192:924–931. doi: 10.1016/j.ajog.2004.11.061. [DOI] [PubMed] [Google Scholar]
- 211.Hirota Y, Cha J, Yoshie M, Daikoku T, Dey SK. Heightened uterine mammalian target of rapamycin complex 1 (mTORC1) signaling provokes preterm birth in mice. Proceedings of the National Academy of Sciences. 2011;108:18073–18078. doi: 10.1073/pnas.1108180108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Mendelson CR, Condon JC. New insights into the molecular endocrinology of parturition. J Steroid Biochem Mol Biol. 2005;93:113–119. doi: 10.1016/j.jsbmb.2004.12.027. [DOI] [PubMed] [Google Scholar]
- 213.Condon JC, Jeyasuria P, Faust JM, Mendelson CR. Surfactant protein secreted by the maturing mouse fetal lung acts as a hormone that signals the initiation of parturition. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:4978–4983. doi: 10.1073/pnas.0401124101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Adams WM, Wagner WC. The role of corticoids in parturition. Biology Report. 1970;3:228. doi: 10.1093/biolreprod/3.2.223. [DOI] [PubMed] [Google Scholar]
- 215.Challis JR, Manchester EL, Mitchell BF, Patrick JE. Activation of adrenal function in fetal sheep by the infusion of adrenocorticotropin to the fetus in utero. Biology of Reproduction. 1982;27:1026–1032. doi: 10.1095/biolreprod27.5.1026. [DOI] [PubMed] [Google Scholar]
- 216.Elovitz MA, Wang Z, Chien EK, Rychlik DF, Phillippe M. A new model for inflammation-induced preterm birth: the role of platelet-activating factor and Toll-like receptor-4. American Journal of Pathology. 2003;163:2103–2111. doi: 10.1016/S0002-9440(10)63567-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Hirsch E, Saotome I, Hirsh D. A model of intrauterine infection and preterm delivery in mice. American Journal of Obstetrics & Gynecology. 1995;172:1598–1603. doi: 10.1016/0002-9378(95)90503-0. [DOI] [PubMed] [Google Scholar]
- 218.Bizargity P, Del Rio R, Phillippe M, Teuscher C, Bonney EA. Resistance to lipopolysaccharide-induced preterm delivery mediated by regulatory T cell function in mice. Biol Reprod. 2009;80:874–881. doi: 10.1095/biolreprod.108.074294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Dombroski RA, Woodard DS, Harper MJK, Gibbs RS. A rabbit model for bacteria-induced preterm pregnancy loss. American Journal of Obstetrics and Gynecology. 1990;163:1938–1943. doi: 10.1016/0002-9378(90)90777-5. [DOI] [PubMed] [Google Scholar]
- 220.Katsuki Y, Kaga N, Kakinuma C, Takagaki K, Kajikawa S, Shibutani Y. Ability of intrauterine bacterial lipopolysaccharide to cause in situ uterine contractions in pregnant rabbits. Acta Obstet Gynecol Scand. 1997;76:26–32. doi: 10.3109/00016349709047780. [DOI] [PubMed] [Google Scholar]
- 221.Davies JK, Shikes RH, Sze CI, Leslie KK, McDuffie RS, Jr, Romero R, Gibbs RS. Histologic inflammation in the maternal and fetal compartments in a rabbit model of acute intra-amniotic infection. American Journal of Obstetrics and Gynecology. 2000;183:1088–1093. doi: 10.1067/mob.2000.108888. [DOI] [PubMed] [Google Scholar]
- 222.Gravett MG. Animal Models for Infection-Induced Preterm Birth. Infection-Induced Preterm Birth in Non-Human Primates. Abstracts of the Interscience Conference on Antimicrobial Agents and Chemotherapy. 2006;46:497. [Google Scholar]
- 223.Adams Waldorf KM, Persing D, Novy MJ, Sadowsky DW, Gravett MG. Pretreatment with toll-like receptor 4 antagonist inhibits lipopolysaccharide-induced preterm uterine contractility, cytokines, and prostaglandins in rhesus monkeys. Reproductive Sciences. 2008;15:121–127. doi: 10.1177/1933719107310992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Gravett MG, Adams KM, Sadowsky DW, Grosvenor AR, Witkin SS, Axthelm MK, Novy MJ. Immunomodulators plus antibiotics delay preterm delivery after experimental intraamniotic infection in a nonhuman primate model. American Journal of Obstetrics & Gynecology. 2007;197:518.e511–518.e518. doi: 10.1016/j.ajog.2007.03.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Yellon SM, Ebner CA, Elovitz MA. Medroxyprogesterone Acetate Modulates Remodeling, Immune Cell Census, and Nerve Fibers in the Cervix of a Mouse Model for Inflammation-induced Preterm Birth. Reproductive Sciences. 2009;16:257–264. doi: 10.1177/1933719108325757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Bytautiene E, Romero R, Vedernikov YP, El-Zeky F, Saade GR, Garfield RE. Induction of premature labor and delivery by allergic reaction and prevention by histamine H1 receptor antagonist. American Journal of Obstetrics & Gynecology. 2004;191:1356–1361. doi: 10.1016/j.ajog.2004.06.092. [DOI] [PubMed] [Google Scholar]
- 227.Bytautiene E, Vedernikov YP, Saade GR, Romero R, Garfield RE. IgE-independent mast cell activation augments contractility of nonpregnant and pregnant guinea pig myometrium. Int Arch Allergy Immunol. 2008;147:140–146. doi: 10.1159/000135701. [DOI] [PubMed] [Google Scholar]
- 228.Kass EH. Effect of corticosteroids and of hormones of pregnancy on the lethal action of bacterial endotoxin bacteriuria and pyelonephritis of pregnancy. Ann N Y Acad Sci. 1960;88:107–115. doi: 10.1111/j.1749-6632.1960.tb20012.x. [DOI] [PubMed] [Google Scholar]
- 229.Henderson M, Entwisle G, Tayback M. Bacteriuria and pregnancy outcome: preliminary findings. Am J Public Health Nations Health. 1962;52:1887–1893. doi: 10.2105/ajph.52.11.1887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Harper MJ, Skarnes RC. Inhibition of abortion and fetal death produced by endotoxin or prostaglandin F2alpha. Prostaglandins. 1972;2:295–309. doi: 10.1016/s0090-6980(72)80017-0. [DOI] [PubMed] [Google Scholar]
- 231.Wren BG. Premature labour with renal infections: the action of coliform endotoxin on the pregnant rat uterus. Australian & New Zealand Journal of Obstetrics & Gynaecology. 1970;10:211–217. doi: 10.1111/j.1479-828x.1970.tb00432.x. [DOI] [PubMed] [Google Scholar]
- 232.Anderson KV, Jurgens G, Nusslein-Volhard C. Establishment of dorsal-ventral polarity in the Drosophila embryo: genetic studies on the role of the Toll gene product. Cell. 1985;42:779–789. doi: 10.1016/0092-8674(85)90274-0. [DOI] [PubMed] [Google Scholar]
- 233.Romero R, Mazor M, Tartakovsky B. Systemic administration of interleukin-1 induces preterm parturition in mice. American Journal of Obstetrics & Gynecology. 1991;165:t-71. doi: 10.1016/0002-9378(91)90450-6. [DOI] [PubMed] [Google Scholar]
- 234.Fidel PL, Jr, Romero R, Wolf N, Cutright J, Ramirez M, Araneda H, Cotton DB. Systemic and local cytokine profiles in endotoxin-induced preterm parturition in mice. American Journal of Obstetrics and Gynecology. 1994;170:1467–1475. doi: 10.1016/s0002-9378(94)70180-6. [DOI] [PubMed] [Google Scholar]
- 235.Medzhitov R, Preston-Hurlburt P, Janeway CA., Jr A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388:394–397. doi: 10.1038/41131. [DOI] [PubMed] [Google Scholar]
- 236.Schwandner R, Dziarski R, Wesche H, Rothe M, Kirschning CJ. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem. 1999;274:17406–17409. doi: 10.1074/jbc.274.25.17406. [DOI] [PubMed] [Google Scholar]
- 237.Yang RB, Mark MR, Gray A, Huang A, Xie MH, Zhang M, Goddard A, Wood WI, Gurney AL, Godowski PJ. Toll-like receptor-2 mediates lipopolysaccharide-induced cellular signalling. Nature. 1998;395:284–288. doi: 10.1038/26239. [DOI] [PubMed] [Google Scholar]
- 238.Qureshi ST, Lariviere L, Leveque G, Clermont S, Moore KJ, Gros P, Malo D. Endotoxin-tolerant mice have mutations in Toll-like receptor 4 (Tlr4) J Exp Med. 1999;189:615–625. doi: 10.1084/jem.189.4.615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Lorenz E, Hallman M, Marttila R, Haataja R, Schwartz DA. Association between the Asp299Gly polymorphisms in the Toll-like receptor 4 and premature births in the Finnish population. Pediatr Res. 2002;52:373–376. doi: 10.1203/00006450-200209000-00011. [DOI] [PubMed] [Google Scholar]
- 240.Elovitz MA, Wang Z, Chien EK, Rychlik DF, Phillippe M. A new model for inflammation-induced preterm birth: the role of platelet-activating factor and Toll-like receptor-4. American Journal of Pathology. 2003;163:2103–2111. doi: 10.1016/S0002-9440(10)63567-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Filipovich Y, Lu SJ, Akira S, Hirsch E. The adaptor protein MyD88 is essential for E coli-induced preterm delivery in mice. Am J Obstet Gynecol. 2009;200:93 e91–93.e98. doi: 10.1016/j.ajog.2008.08.038. [DOI] [PubMed] [Google Scholar]
- 242.Blank V, Hirsch E, Challis JRG, Romero R, Lye SJ. Cytokine signaling, inflammation, innate immunity and preterm labour - a workshop report. Placenta. 2008;29(Suppl A):S102–S104. doi: 10.1016/j.placenta.2007.10.011. [DOI] [PubMed] [Google Scholar]
- 243.Lin Y, Xie M, Chen Y, Di J, Zeng Y. Preterm delivery induced by LPS in syngeneically impregnated BALB/c and NOD/SCID mice. Journal of Reproductive Immunology. 2006;71:87–101. doi: 10.1016/j.jri.2006.01.005. [DOI] [PubMed] [Google Scholar]
- 244.Wang H, Hirsch E. Bacterially-induced preterm labor and regulation of prostaglandin-metabolizing enzyme expression in mice: the role of toll-like receptor 4. Biology of Reproduction. 2003;69:1957–1963. doi: 10.1095/biolreprod.103.019620. [DOI] [PubMed] [Google Scholar]
- 245.Beijar EC, Mallard C, Powell TL. Expression and subcellular localization of TLR-4 in term and first trimester human placenta. Placenta. 2006;27:322–326. doi: 10.1016/j.placenta.2004.12.012. [DOI] [PubMed] [Google Scholar]
- 246.Kim YM, Romero R, Chaiworapongsa T, Kim GJ, Kim MR, Kuivaniemi H, Tromp G, Espinoza J, Bujold E, Abrahams VM, Mor G. Toll-like receptor-2 and -4 in the chorioamniotic membranes in spontaneous labor at term and in preterm parturition that are associated with chorioamnionitis. American Journal of Obstetrics & Gynecology. 2004;191:1346–1355. doi: 10.1016/j.ajog.2004.07.009. [DOI] [PubMed] [Google Scholar]
- 247.Hirschfeld AF, Jiang R, Robinson WP, McFadden DE, Turvey SE. Toll-like receptor 4 polymorphisms and idiopathic chromosomally normal miscarriage. Human Reproduction. 2007;22:440–443. doi: 10.1093/humrep/del377. [DOI] [PubMed] [Google Scholar]
- 248.Ilievski V, Hirsch E. Synergy between viral and bacterial toll-like receptors leads to amplification of inflammatory responses and preterm labor in the mouse. Biol Reprod. 2010;83:767–773. doi: 10.1095/biolreprod.110.085464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Koga K, Cardenas I, Aldo P, Abrahams VM, Peng B, Fill S, Romero R, Mor G. Activation of TLR3 in the trophoblast is associated with preterm delivery. Am J Reprod Immunol. 2009;61:196–212. doi: 10.1111/j.1600-0897.2008.00682.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Abrahams VM, Aldo PB, Murphy SP, Visintin I, Koga K, Wilson G, Romero R, Sharma S, Mor G. TLR6 modulates first trimester trophoblast responses to peptidoglycan. Journal of Immunology. 2008;180:6035–6043. doi: 10.4049/jimmunol.180.9.6035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Cardenas I, Mor G, Aldo P, Lang SM, Stabach P, Sharp A, Romero R, Mazaki-Tovi S, Gervasi M, Means RE. Placental viral infection sensitizes to endotoxin-induced pre-term labor: a double hit hypothesis. Am J Reprod Immunol. 2011;65:110–117. doi: 10.1111/j.1600-0897.2010.00908.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Scharfe-Nugent A, Corr SC, Carpenter SB, Keogh L, Doyle B, Martin C, Fitzgerald KA, Daly S, O'Leary JJ, O'Neill LAJ. TLR9 provokes inflammation in response to fetal DNA: mechanism for fetal loss in preterm birth and preeclampsia. Journal of Immunology. 2012;188:5706–5712. doi: 10.4049/jimmunol.1103454. [DOI] [PubMed] [Google Scholar]
- 253.Inder T, Neil J, Yoder B, Rees S. Patterns of cerebral injury in a primate model of preterm birth and neonatal intensive care. Journal of Child Neurology. 2005;20:965–967. doi: 10.1177/08830738050200120601. [DOI] [PubMed] [Google Scholar]
- 254.Romero R, Chaiworapongsa T, Kuivaniemi H, Tromp G. Bacterial vaginosis, the inflammatory response and the risk of preterm birth: a role for genetic epidemiology in the prevention of preterm birth. American Journal of Obstetrics & Gynecology. 2004;190:1509–1519. doi: 10.1016/j.ajog.2004.01.002. [comment]. [DOI] [PubMed] [Google Scholar]
- 255.Kim YM, Romero R, Chaiworapongsa T, Espinoza J, Mor G, Kim CJ. Dermatitis as a component of the fetal inflammatory response syndrome is associated with activation of Toll-like receptors in epidermal keratinocytes. Histopathology. 2006;49:506–514. doi: 10.1111/j.1365-2559.2006.02542.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Bell MJ, Hallenbeck JM, Gallo V. Determining the fetal inflammatory response in an experimental model of intrauterine inflammation in rats. Pediatric Research. 2004;56:541–546. doi: 10.1203/01.PDR.0000139407.89883.6B. [DOI] [PubMed] [Google Scholar]
- 257.Rees SM, Duncan J, Loeliger M, Cock M, Harding R. Repeated exposure to bacterial endotoxin during gestation leads to cerebral white matter and retinal damage in the ovine fetus. Society for Neuroscience Abstracts. 2001;27:2406. [Google Scholar]
- 258.Saadani-Makki F, Kannan S, Lu X, Janisse J, Dawe E, Edwin S, Romero R, Chugani D. Intrauterine administration of endotoxin leads to motor deficits in a rabbit model: a link between prenatal infection and cerebral palsy. American Journal of Obstetrics & Gynecology. 2008;199:651.e651–651.e657. doi: 10.1016/j.ajog.2008.06.090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Burd I, Chai J, Gonzalez J, Ofori E, Monnerie H, Le Roux PD, Elovitz MA. Beyond white matter damage: fetal neuronal injury in a mouse model of preterm birth. American Journal of Obstetrics & Gynecology. 2009;201:279–278. doi: 10.1016/j.ajog.2009.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Breen K, Brown A, Burd I, Chai J, Friedman A, Elovitz MA. TLR-4-dependent and -independent mechanisms of fetal brain injury in the setting of preterm birth. Reproductive Sciences. 2012;19:839–850. doi: 10.1177/1933719112438439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Elovitz MA, Mrinalini C, Sammel MD. Elucidating the early signal transduction pathways leading to fetal brain injury in preterm birth. Pediatric Research. 2006;59:50–55. doi: 10.1203/01.pdr.0000191141.21932.b6. [DOI] [PubMed] [Google Scholar]
- 262.Robertson SA, Skinner RJ, Care AS. Essential role for IL-10 in resistance to lipopolysaccharide-induced preterm labor in mice. Journal of Immunology. 2006;177:4888–4896. doi: 10.4049/jimmunol.177.7.4888. [DOI] [PubMed] [Google Scholar]
- 263.Thaxton JE, Romero R, Sharma S. TLR9 activation coupled to IL-10 deficiency induces adverse pregnancy outcomes. Journal of Immunology. 2009;183:1144–1154. doi: 10.4049/jimmunol.0900788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Dordelmann M, Kerk J, Dressler F, Brinkhaus MJ, Bartels DB, Dammann CEL, Dork T, Dammann O. Interleukin-10 high producer allele and ultrasound-defined periventricular white matter abnormalities in preterm infants: a preliminary study. Neuropediatrics. 2006;37:130–136. doi: 10.1055/s-2006-924554. [DOI] [PubMed] [Google Scholar]
- 265.Barker DJP. In utero programming of cardiovascular disease. Theriogenology. 2000;53:555–574. doi: 10.1016/s0093-691x(99)00258-7. [DOI] [PubMed] [Google Scholar]
- 266.Barker DJ, Gluckman PD, Godfrey KM, Harding JE, Owens JA, Robinson JS. Fetal nutrition and cardiovascular disease in adult life. Lancet. 1993;341:938–941. doi: 10.1016/0140-6736(93)91224-a. [DOI] [PubMed] [Google Scholar]
- 267.Seki Y, Williams L, Vuguin PM, Charron MJ. Minireview: Epigenetic programming of diabetes and obesity: animal models. Endocrinology. 2012;153:1031–1038. doi: 10.1210/en.2011-1805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Long NM, Ford SP, Nathanielsz PW. Multigenerational effects of fetal dexamethasone exposure on the hypothalamic-pituitary-adrenal axis of first and second generation female offspring. Am J Obstet Gynecol. 2012;6:014. doi: 10.1016/j.ajog.2012.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Okada Y, Ueshin Y, Isotani A, Saito-Fujita T, Nakashima H, Kimura K, Mizoguchi A, Oh-hora M, Mori Y, Ogata M, Oshima RG, Okabe M, Ikawa M. Complementation of placental defects and embryonic lethality by trophoblast-specific lentiviral gene transfer. Nat Biotech. 2007;25:233–237. doi: 10.1038/nbt1280. [DOI] [PubMed] [Google Scholar]
- 270.Kodama K, Horikoshi M, Toda K, Yamada S, Hara K, Irie J, Sirota M, Morgan AA, Chen R, Ohtsu H, Maeda S, Kadowaki T, Butte AJ. Expression-based genome-wide association study links the receptor CD44 in adipose tissue with type 2 diabetes. Proc Natl Acad Sci U S A. 2012;109:7049–7054. doi: 10.1073/pnas.1114513109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Hou ZC, Sterner KN, Romero R, Than NG, Gonzalez JM, Weckle A, Xing J, Benirschke K, Goodman M, Wildman DE. Elephant transcriptome provides insights into the evolution of eutherian placentation. Genome Biol Evol. 2012;4:713–725. doi: 10.1093/gbe/evs045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Shorten PR, Peterson AJ, O’Connell AR, Juengel JL, McNatty KP, Soboleva TK. A mathematical model of pregnancy recognition in mammals. Journal of Theoretical Biology. 2010;266:62–69. doi: 10.1016/j.jtbi.2010.06.005. [DOI] [PubMed] [Google Scholar]
- 273.Williams PH, Eyles R, Weiller G. Plant MicroRNA Prediction by Supervised Machine Learning Using C5.0 Decision Trees. J Nucleic Acids. 2012;2012:652979. doi: 10.1155/2012/652979. [DOI] [PMC free article] [PubMed] [Google Scholar]