Skip to main content
American Journal of Physiology - Heart and Circulatory Physiology logoLink to American Journal of Physiology - Heart and Circulatory Physiology
. 2024 May 17;327(1):H191–H220. doi: 10.1152/ajpheart.00055.2024

Guidelines for assessing maternal cardiovascular physiology during pregnancy and postpartum

Helen E Collins 1,, Barbara T Alexander 2, Alison S Care 3, Margie H Davenport 4, Sandra T Davidge 4, Mansoureh Eghbali 5, Dino A Giussani 6, Martijn F Hoes 7, Colleen G Julian 8, Holly A LaVoie 9, I Mark Olfert 10, Susan E Ozanne 6, Egle Bytautiene Prewit 11, Junie P Warrington 2, Lubo Zhang 12, Styliani Goulopoulou 12,
PMCID: PMC11380979  PMID: 38758127

Abstract

Maternal mortality rates are at an all-time high across the world and are set to increase in subsequent years. Cardiovascular disease is the leading cause of death during pregnancy and postpartum, especially in the United States. Therefore, understanding the physiological changes in the cardiovascular system during normal pregnancy is necessary to understand disease-related pathology. Significant systemic and cardiovascular physiological changes occur during pregnancy that are essential for supporting the maternal-fetal dyad. The physiological impact of pregnancy on the cardiovascular system has been examined in both experimental animal models and in humans. However, there is a continued need in this field of study to provide increased rigor and reproducibility. Therefore, these guidelines aim to provide information regarding best practices and recommendations to accurately and rigorously measure cardiovascular physiology during normal and cardiovascular disease-complicated pregnancies in human and animal models.

Keywords: animal models, cardiac physiology, comorbidities, hypertensive disorders of pregnancy, vascular function

INTRODUCTION

Pregnancy is a unique physiological stress test that many women will experience within their lifetime. Despite this, there is little understanding of how the cardiovascular system adapts during uncomplicated, healthy pregnancies, with greater emphasis placed on understanding and reducing the pregnancy-associated cardiovascular disease burden. With maternal mortality rates across the world at an all-time high and likely to increase further in subsequent years, having a complete understanding of these cardiovascular adaptations is imperative. To adequately advance the field and to effectively aid in reducing maternal mortality rates and designing rational interventions, a greater understanding is needed concerning the plethora of cardiovascular physiological changes that occur during both normal and cardiovascular disease-complicated pregnancies.

Normal, uncomplicated pregnancies are characterized by several hormonal, systemic, and cardiovascular adaptations necessary to support maternal-fetal circulation and, ultimately, a healthy pregnancy. Understanding of these critical cardiovascular adaptations has been hindered by the lack of experimental consistency among studies and, in some cases, a lack of rigor in reporting key biological variables that impact pregnancy, postpartum, and lactation. Here, we review the best practices and recommendations to accurately and rigorously measure cardiovascular physiology during normal pregnancies and those complicated by cardiovascular disease in human and animal models. We discuss key physiological changes in the cardiovascular system that have been documented in both human and animal models and preferred experimental techniques used to examine these changes in vivo and in vitro, i.e., “pregnancy in a dish.” We highlight key pregnancy-associated variables that should be considered during experimental design, such as gravidity, parity, species used, and selection of time points or periods to intervene during pregnancy. In addition, we discuss the impact of several modifiable stressors of increasing importance, such as the impact of changes in diet, physical activity, and environmental exposures. We finally discuss the advantages and disadvantages of current models of pregnancy-associated cardiovascular diseases, such as hypertensive disorders of pregnancy, myocardial infarction, and peripartum cardiomyopathy, and provide recommendations for their respective uses. After increasing rigor and reproducibility, a secondary goal of these guidelines is to provide a framework for investigators to understand preferred methodologies and necessary considerations for completing meaningful assessments of maternal cardiovascular physiology.

Although this article focuses on assessing maternal cardiovascular physiology during pregnancy and postpartum, investigators with a combined interest in developmental programming should refer to our paired article “Guidelines for In Vivo Models of Developmental Programming of Cardiovascular Disease Risk.”

OVERVIEW OF MATERNAL CARDIOVASCULAR PHYSIOLOGY DURING PREGNANCY

Significant changes in systemic and cardiovascular physiology occur during pregnancy and postpartum, facilitating the physiological demands of a healthy pregnancy, labor and delivery, and postpartum recovery. In particular, there is a ∼48% expansion in plasma volume and a ∼30–40% increase in cardiac output (through increased stroke volume in rodents) above nonpregnant levels to facilitate the increasing metabolic demands of the growing fetus and a new organ, the placenta (13). Significant changes in sympathetic and parasympathetic balance during pregnancy contribute to reductions in heart rate variability during early pregnancy (4). A reduction in total peripheral resistance (∼35–40%) coupled with an increase in vascular compliance (1) prevents an increase in arterial pressure, which shows a transient reduction in midgestation in healthy pregnant humans, sheep, and rodents (58). Additional changes in maternal physiology include changes in maternal metabolism, necessary to divert nutrients to the growing fetus (2) (highlighted in Fig. 1). Our intent in this section is to briefly introduce the main maternal cardiac and vascular changes that occur during pregnancy. The reader is directed to comprehensive reviews that discuss in detail these adaptations (2, 6, 9, 10).

Figure 1.

Figure 1.

Systemic and cardiovascular adaptations during pregnancy. A schematic depicting the key changes in both systemic and cardiovascular physiology that occur in pregnant women and animals. These adaptations must occur to meet the circulatory demands of the mother and fetus. Several hemodynamic changes occur including changes in cardiac output (CO), blood pressure (BP), and total peripheral resistance (TPR). In addition, there is significant remodeling of the ventricles, atria, aorta, and the vasculature, with the latter including remodeling of and increased blood flow to the uterine arteries. In addition, the placenta develops and provides vital nutrients necessary to sustain fetal growth and development, assisted by changes in maternal systemic metabolism. Arrows depict key systemic and cardiovascular changes common among all of the depicted mammalian species. Images were created with a licensed version of BioRender.com.

Cardiac Adaptations

The increased workload placed on the heart during pregnancy results in the physiological growth of the heart, i.e., an increase in heart size and mass and an increase in left ventricular chamber dimensions. The increase in heart size during pregnancy represents physiological hypertrophy that is thought to be reversible and occurs independent of changes associated with pathological hypertrophy, such as fibrosis, reduced cardiac function, and decreased angiogenesis (11, 12). However, this physiological hypertrophy is associated with temporal changes in different cardiac cell compartments and significant changes in cardiac glucose, lipid, and ketone body metabolism (1315). Many maternal cardiovascular responses to stress are also diminished. For instance, the maternal chronotropic response to hypoxia is markedly reduced (16). This highlights that pregnancy not only affects basal but also stimulates cardiovascular function in the mother (16).

Many preclinical pregnancy studies have used commonly used experimental techniques to assess changes in systemic physiology and metabolism, such as the assessment of circulating factors and metabolites in the blood, through glucose tolerance tests, insulin tolerance tests, and evaluation of key metabolic organs. Studies primarily focused on the cardiac adaptations during a healthy pregnancy comprise standard documentation of mouse cardiac physiology (1113, 1719). In recent years, studies have also used omics approaches for interrogation of maternal systemic and cardiovascular adaptations to normal pregnancy (13); however, many have focused mostly on pathological consequences.

Vascular Adaptations

The maternal vascular system undergoes significant adaptations during pregnancy (9). Vascular adaptations occur at the cellular level, determining the function of the main components of the vascular wall and, consequently, facilitating changes in vascular tone and reactivity to circulating and local factors. During pregnancy, endothelial cells increase the production and bioavailability of vasodilatory products such as nitric oxide, accommodate large increases in fluid volumes, and form gap junctions in the uterine circulation that enhance vasodilatory responses (2022). One of the primary changes in vascular smooth muscle cell function during pregnancy is a change in myogenic tone, primarily in uterine arteries (2325). In pregnant women, the myogenic tone of myometrial arteries increases in late pregnancy (26), whereas uterine arteries from pregnant mice have reduced myogenic tone compared with nonpregnant mice (27). Perivascular adipose tissue also changes during pregnancy and this change is vascular bed specific. Adipose tissue surrounding rodent mesenteric arteries becomes anticontractile (28), whereas uterine perivascular adipose tissue has antidilatory effects that are mediated by distinct mechanisms in pregnant versus nonpregnant rats (29, 30).

The most pronounced changes in the maternal vascular system during pregnancy occur in the uteroplacental circulation, where hemodynamic adaptations continue a pattern established during the menstrual cycle (31). This pattern involves a reduction in uterine artery impedance and increased uterine artery blood flow, which reaches a >10-fold increase at term compared with prepregnancy levels (32). Extensive growth of the uterine circulation is a hallmark of a healthy pregnancy that facilitates large changes in uterine blood flow and the establishment of blood flow to a newly developed organ, the placenta. In humans and rodents, the placenta is hemochorial, and this anatomical architecture provides a limited barrier to the delivery of nutrients from the mother to the fetus (33). To support an increase in uterine and placental blood flow, uterine arteries, and veins experience outward expansive remodeling and an increase in length (8, 32). In species with hemochorial placentation, remodeling of the uterine spiral arteries, which are nonbranching end arteries that supply the outer endometrial layer (or decidua in pregnancy), contributes to a reduction in downstream resistance, secondary to the placentation process, and acceleration of the arterial blood in upstream vessels (i.e., ascending uterine arteries, arcuate, and radial arteries). This hemodynamic adaptation increases shear stress on the vascular wall and shear stress-induced production of nitric oxide from the vascular endothelium (10). Remodeling and increased dilatory responses of the uterine arteries, as well as local changes in cell signaling and altered responsiveness to paracrine and humoral factors, further contribute to pregnancy-induced increases in uterine blood flow (8, 3440). The time course of vascular remodeling differs between small/preplacental uterine vessels and upstream large uterine vessels. Changes in large vessels start early in pregnancy (41) and may occur independently of the presence of the placenta (42). Pregnancy-associated adaptations of the uterine circulation differ among mammals, and this variation is due to differences in structure and anatomy of the uterine circulation, type of placentation [i.e., hemochorial in humans vs. epitheliochorial in ruminants or endotheliochorial in dogs (43)], uterine size, and number of offspring (44).

In addition to uterine blood flow, splanchnic (e.g., stomach, spleen, liver, intestines, and pancreas), renal, pulmonary, and skin blood flow increases during pregnancy (45). In the cerebral circulation, receptor, and transporter activity changes, and the upper and lower limits of the cerebral blood flow autoregulatory curve extend during pregnancy, maintaining brain homeostasis and protecting the mother’s brain from blood pressure fluctuations (46). Despite these alterations, the changes in cerebral blood flow during pregnancy are minimal (46). The fetoplacental unit secretes signaling molecules into the maternal circulation that can act upon cardiac and vascular cells to induce functional and structural changes. Thus, fetoplacental signals play a key role in the expansion and adaptability of the maternal cardiovascular system during pregnancy [for detailed discussion, see Osol et al. (9)].

Postpartum Cardiovascular Adaptations

Little is known about the temporal changes of the maternal vascular system during the immediate or long-term postpartum period following a healthy pregnancy. Mean arterial pressure is reduced, and arterial compliance is increased one year postpartum compared with prepregnancy values in women who had a healthy pregnancy, suggesting a long-lasting beneficial effect of pregnancy on the female vascular system (47). Pregnancy-induced remodeling of the uterine arteries is partially reversed during the postpartum period, whereas remodeling of arteries in nonvascular beds, such as the ascending aorta, is fully reversed during postpartum (47).

BEST PRACTICES FOR STUDYING MATERNAL CARDIOVASCULAR PHYSIOLOGY DURING PREGNANCY AND POSTPARTUM

An examination of recent American Journal of Physiology articles in the past 10 years has shown that there appears to be a significant discrepancy regarding reporting key biological and pregnancy-related variables within animal studies. For example, between published articles, there seems to be a lack of consistency between the specific time points examined during pregnancy and postpartum, alongside a lack of information or consistency regarding essential study information, such as the starting age and particular strain of animals used (e.g., C57BL/6 mice; 6 N or 6 J), and pregnancy-related information such as litter sizes, number of pregnancies, gestational length, lactation status, and hormonal status. In addition, sometimes, it is unclear whether the primary focus of the studies is maternal, fetal, or both. In some cases, the key cardiovascular adaptations are not always fully assessed. These considerations will be discussed in further detail in the following sections.

Studying Cardiovascular Physiology of Pregnancy in Human Subjects

The rapid and profound physiological changes of pregnancy significantly affect the conduct and interpretation of standard cardiovascular assessments. Adaptations are progressive but occur at different time lines and trajectories during and following pregnancy (i.e., blood pressure vs. heart rate) (2). Thus, it is important to standardize assessments based on gestational age and time since childbirth when conducting cardiovascular assessments during the peripartum period. The selection of the appropriate control group will be dependent on the primary research question, and whenever possible control or match groups for key population characteristics that are known to influence cardiovascular health [e.g., age, body mass index (BMI), parity, gestational age]. For example, if the primary study aim was to examine the impact of preeclampsia on cardiovascular health, the control group would consist of pregnant women not diagnosed with preeclampsia who were matched primarily by gestational age and, as much as possible, age, parity, and BMI. In contrast, if the primary aim was to examine the impact of pregnancy on cardiovascular health, the control group would be matched by key population characteristics (e.g., age, BMI, parity).

Key considerations for cardiovascular testing during pregnancy.

  • 1) 

    Determine (and report) weeks gestation, gravidity, and parity (2).

  • 2) 

    Screen for a prior or current history of pregnancy complications known to impact cardiovascular health (e.g., gestational diabetes, gestational hypertension, preeclampsia; emerging risk factors include preterm delivery, intrauterine growth restriction) (48).

  • 3) 

    Understand the normal and abnormal physiological adaptations to pregnancy so that medical referral can be made, as appropriate, if observed (e.g., undiagnosed hypertension, hyperglycemia) (49).

  • 4) 

    Follow up with the mother postchildbirth to identify pregnancy complications that may have developed later in/after pregnancy, as there is some evidence that cardiovascular dysfunction can be present before diagnosis of complications (e.g., preeclampsia) (50, 51).

  • 5) 

    Normal pretest standardization for cardiovascular testing in nonpregnant populations should be followed in pregnant and postpartum populations (e.g., fasted >6 h, avoid exercise >24 h, no intake of caffeine, vitamin C, alcohol, and smoking for >12 h) (52). However, because of pregnancy-induced changes in metabolism and glucose stores, lengthy fasted protocols may not be advisable in some pregnant populations (e.g., insulin-treated gestational diabetes mellitus). When fasted protocols cannot be conducted, standardized pretest nutritional intake is recommended (e.g., a standard snack rationed by grams of macronutrients per kilogram body weight at a prespecified interval before the test). It is, however, recognized that this is not always feasible.

  • 6) 

    The time of day that the vascular assessment was conducted should be standardized and reported to account for diurnal changes in cardiovascular function (52).

  • 7) 

    As pregnancy progresses, the uterus may compress the aorta and vena cava in the supine position leading to transient alterations in venous return, cardiac output, blood pressure, and heart rate (49). The supine position should be avoided in favor of a left lateral tilt in the recumbent position, or a semirecumbent position when measurements are being made.

  • 8) 

    When longitudinal cardiovascular assessments are conducted, maternal posture should be maintained across all assessments from preconception to postpartum.

  • 9) 

    When feasible, blood sampling to assess sex hormones (estrogen, progesterone, and testosterone) (49), glucose, lipid, and insulin is recommended given their known influence on cardiovascular health and function.

Although many physiological adaptations reverse quickly after childbirth, others remain transiently (or permanently) altered. Some of the cardiovascular adaptations to pregnancy may persist past the first-year postpartum (e.g., increased cardiac output, aortic diameter) thus postpartum “controls” should not be considered the same as individuals who have never been pregnant. Breastfeeding may also impact cardiovascular health (53). Thus, data on the length and type of breastfeeding (e.g., exclusive vs. mixed) should be recorded.

Modeling Cardiovascular Physiology of Pregnancy in Experimental Animals

There are many mammalian species that can be used to model human pregnancy, each with important strengths and weaknesses (see Table 1). Genetically modified mice are a powerful tool that can be used to understand molecular mechanisms underlying maternal cardiovascular health and disease as a result of their similarity to humans, short gestation period, and the emergence of genomics (54). In mice (55), rats (56), rabbits (57), and guinea pigs (58), the maternal cardiovascular adaptations to pregnancy can be studied noninvasively, for instance focusing on measurements of maternal cardiac geometry and function using echocardiography, or calculating pulsatility index (PI, the measure of blood flow velocity in a vessel during the cardiac cycle), as a marker of uteroplacental vascular resistance via Doppler blood flow velocimetry. In addition, although used less frequently, cardiac magnetic resonance imaging has been used to assess cardiac function in monkey pregnancy studies (59). In the smaller species, echocardiography is usually performed under reversible anesthesia and, therefore, can be done serially. However, care must be taken because anesthesia can only be induced a fixed number of times in any one animal, and the anesthetic itself may depress maternal cardiovascular function (60). Radiotelemetry in rodents circumvents some of these problems, but cardiovascular measurements obtained using this method are currently limited to changes in resting blood pressure and heart rate (61). A few studies have attempted chronically instrumented preparations in rats surgically prepared with arterial and venous catheters and a femoral Transonic flow probe (see Table 1). Following postsurgical recovery, drugs can be administered to study not only basal but also stimulated cardiovascular function (6264). In mice, rats, rabbits, and guinea pigs, tissues can also be isolated at terminal endpoints. One can study molecular mechanisms in the placenta (65, 66) and cardiovascular function in the pregnant and postpartum animal via isolated Langendorff preparations (67, 68), and perfusion or wire myography (69, 70). However, some rodent species present important weaknesses, which need careful consideration. Rats are a better model for studying placentation due to the presence of hemochorial placentation, observed in both humans and rats (71). However, in rats and mice, cardiovascular maturation continues past birth, becoming complete by the second week of postnatal life (72). Rodents also give birth to litters, so there may be differences in the maternal metabolic adaptation to pregnancy. Smaller species will also have low blood volume, limiting the number of blood samples that could be taken serially, for instance, with advancing gestation. In contrast, sheep and humans share similar tempos of prenatal cardiovascular development (72) and some breeds of sheep, like Welsh Mountain, give birth primarily to singleton lambs of similar weight to term human babies (7). Most importantly, the sheep permits surgical instrumentation of the mother and fetus for long-term in vivo cardiovascular recording and serial blood sampling for endocrinology and daily monitoring of blood gases and metabolic status (7, 7376). Recently, there has been an important development for research in sheep with the introduction of CamDAS technology (shown in Fig. 2). CamDAS is a wireless data acquisition system that permits the simultaneous recording of multiple pressure and blood flow signals from mother and fetus in vivo over long periods of gestation in free-moving animals (7, 7376). The systems not only improve animal welfare and, thereby, the physiological quality of the recording, but they also permit the continuous measurement of cardiovascular function in the mother and the fetus beat-to-beat in different environments for weeks to months. For instance, by combining CamDAS technology with the creation of hypoxic isolators, it has been possible to investigate longitudinally the effect of hypoxic pregnancy on the maternal cardiovascular adaptation to pregnancy and the fetal physiology (7, 7376). Clearly, there are gross anatomical differences between the human hemochorial and the ovine cotyledonary placenta (77). However, there are also important similarities. Both sheep and humans have placental countercurrent flow of maternal and fetal blood within the placental villous tree, comparable transplacental oxygen gradients and oxygen consumption rates, as well as similar nutrient transporter expression (7). At the molecular level, the induction of oxidative and endoplasmic reticulum (ER) stress and the activation of the unfolded protein response (UPR) are highly conserved pathways across species (78). This is also the case for rodents, in which activation of the placental UPRER has been demonstrated in hypoxic pregnancy (79). Llamas, which share similar advantages to sheep and are amenable to similar experimentation, have been used to model human adaptation to high-altitude pregnancy (80). Nonhuman primates like macaques and baboons are clearly the most similar to humans. However, clear disadvantages of the larger and more sentient species, including sheep, llamas, and nonhuman primates, are ethical concerns and high-maintenance costs. For this reason, study of maternal cardiovascular function in nonhuman primate species have been mostly noninvasive in multiuser colonies that are kept very long term (81). Additional examples of examination of maternal cardiovascular physiology are listed in Table 1 (8298).

Table 1.

Modeling human pregnancy in different animal species: comparative advantages and disadvantages in the study of maternal cardiovascular physiology

Pros and Cons
Possible Measurable Outcomes
Species Main Advantages Main Disadvantages Echo/Doppler Serial Blood Sampling In Vivo Chronically Instrumented Preparation Langendorff Preparation Perfusion/ Wire Myography Molecular Stereology/Histology
Mouse Genetic manipulation (PMID: 35510543)
Short gestation; amenable to transgenerational studies
Hemomonochorial placenta (ISBN:978-3-540-78796-9)
Litter bearing
Born immature; does not mirror the tempo of human development (PMID: 35534925).
Small; limited blood volume

Under anesthesia
PMID: 30293577
×
Not reported
Limited
PMIDs: 27927648, 24935937, 22014504, 20100997, 11242593
Limited
PMIDs: 24705556, 25051449

PMIDs: 35510543, 27181166, 26110512

PMIDs: 35510543, 30293577, 25051449, 26110512

PMID: 26110512
Rat Larger than mouse
Short gestation; amenable to transgenerational studies
Hemomonochorial placenta (ISBN:978-3-540-78796-9)
Litter bearing
Born immature; does not mirror the tempo of human development (PMID: 35534925)
Small; limited blood volume

Under anesthesia
PMID: 37506535
×
Not reported
Limited
PMIDs: 23856654, 33788974, 34757774

PMIDs: 33788974, 30354714

PMIDs: 15326063, 37615097, 34694887, 34634151

PMIDs: 33788974, 30248337, 22289909, 34694887, 34634151

PMIDs: 30248337, 22289909
Guinea pig Mirrors tempo of human development (PMIDs: 29633280, 35534925)
Midrange gestation; amenable to transgenerational studies
Hemomonochorial placenta ISBN:978-3-540-78796-9
Litter bearing
Small; limited blood volume

Under anesthesia
PMID: 29633280
×
Not reported
Limited
PMID: 29847165

PMID: 36487188

PMID: 27739590

PMID: 29633280

PMID: 29633280
Rabbit Short gestation; amenable to transgenerational studies
Hemomonochorial placenta
ISBN:978-3-540-78796-9
Litter bearing
Born immature; does not mirror the tempo of human development
PMID: 35534925
Small; limited blood volume

Under anesthesia
PMID: 35927671
×
Not reported
×
Not reported

PMID: 12271152

PMID: 26806799

PMID: 35927671

PMID: 35927671
Sheep Mainly singleton-twin bearing
Mirrors tempo of human development
PMID: 35534925
Large; high blood volume
Cotyledonary placenta
ISBN:978-3-540-78796-9
Expensive

Under anesthesia
PMID: 35534925

PMIDs: 35534925, 31039691, 26926316, 32862711, 34694887

PMIDs: 35534925, 31039691, 26926316, 32862711, 34694887

PMID: 26806799

PMID: 34694887

PMIDs: 35534925, 34694887

PMIDs: 35534925, 34694887
Llama Mainly singleton-twin bearing
Large; high blood volume
Epitheliochorial placenta
ISBN:978-3-540-78796-9
Expensive

Under anesthesia
PMID: 11262476

PMIDs: 12855051, 18006479

PMIDs: 12855051, 18006479
×
Not reported

PMIDs: 12855051, 18006479

PMIDs: 12855051, 18006479

PMIDs: 12855051, 18006479
Nonhuman primate Mainly singleton-twin bearing
Mirrors tempo of human development
PMID:35534925
Large; high blood volume
Hemomonochorial placenta ISBN:978-3-540-78796-9
Expensive
Under anesthesia
PMID: 21447636

PMID: 9546793
×
PMID: 8633635
×
Not reported

PMID: 11262476

PMIDs: 21447636, 9546793

PMIDs: 21447636, 9546793

Figure 2.

Figure 2.

The CamDAS system used for recording maternal and fetal cardiovascular function during hypoxic pregnancy. A: a specially designed nitrogen-generating system supplies compressed air and nitrogen to bespoke isobaric hypoxic chambers housed at The Barcroft Centre, University of Cambridge. Each chamber is equipped with an electronic servo-controlled humidity cool steam injection system to return the appropriate humidity to the inspirate (i). Ambient partial pressures of oxygen and carbon dioxide, humidity, and temperature within each chamber were monitored via sensors (ii). For experimental procedures, each chamber had a double transfer port (iii) to internalize material and a manually operated sliding panel (iv) to bring the ewe into a position, where daily sampling of blood could be achieved through glove compartments (v). Each chamber incorporated a drinking bowl on continuous water supply and a rotating food compartment (vi) for determining food intake. A sealed transfer isolation cart could be attached to a side exit (vii) to couple chambers together for cleaning. Waste could be disposed via a sealable pipe (viii). B: some ewes can be instrumented with the CamDAS system during surgery under general anesthesia. Following postsurgical recovery (usually 5 days), the system can monitor continuous longitudinal changes in maternal and fetal cardiovascular function, for example: maternal and fetal arterial blood pressure, maternal and fetal heart rate and maternal and fetal femoral blood flow, uterine blood flow, umbilical blood flow, and fetal carotid blood flow. The wireless CamDAS system is contained in two parts in a custom-made sheep jacket: the data acquisition box (ix) on one side and a box containing the pressure transducers (x) on the other side. Cables (xi) provide connection between the two boxes and to two lithium battery packs. Measurements made using the CamDAS system are transmitted wirelessly via Bluetooth technology (xiii) to a laptop on the outside (xii), on which it is possible to continuously measure and record the maternal and fetal cardiovascular function during the experimental period. CC-BY: reproduced with permission (7, 7375).

Studying Cardiovascular Physiology of Pregnancy in a Dish: In Vitro Studies

Pregnancy is a multifaceted physiological event affecting numerous organ systems. Studying its molecular pathways in the cardiovascular system in humans is challenging because of required invasive techniques, leading researchers to often use animal models. However, because these models do not fully capture human molecular physiology, consideration of in vitro models for examining pregnancy-related effects should be made in addition to in vivo models. Human pluripotent stem cells, derived from embryonic stem cells or sources like dermal fibroblasts or peripheral blood mononuclear cells (PBMCs), serve as a primary resource. These cells can be studied in homogeneous cultures (99), cocultures (100, 101), or engineered tissues (102, 103).

The systemic nature of pregnancy necessitates the incorporation of various associated factors into these models, including circulating profiles like hormones (104), metabolic substrates (105107), and hemodynamic changes such as heart rate alterations (108) and plasma volume shifts (109). Integrating these factors can be intricate, especially when combining multiple elements. These parameters also change as pregnancy progresses and should be modeled in relation to the stage of pregnancy to be studied.

In vitro heart rate elevation can be achieved via chronic adrenergic stimulation (110, 111) or electrical pacing (112114). Electrical pacing offers a sustained response but can be detrimental to cardiomyocytes over time. In contrast, the efficacy of adrenergic stimulation may diminish because of receptor desensitization. While blood pressure sees a minor rise during normal pregnancy (108), it can be modeled in vitro (i.e., as increased preload) by applying cyclic mechanical stretching (115, 116). Modeling increased cardiac afterload in monolayer cultures is problematic due to cell culture substrate rigidity. Engineered cardiac tissues provide a viable method for simulating preload and afterload, as evidenced in earlier studies (102, 117). To model increased afterload, one can embed the tissue within a framework of rigid pillars that maintain the tissue’s dimensions at its natural resting state. Conversely, preload simulation can be achieved by attaching the tissue to a pliable strip that exerts a pulling force, thereby extending the tissue beyond its resting length.

Metabolic substrate and hormone changes during pregnancy are well documented (105107), allowing culture media adjustments accordingly. Incorporating these hormones at appropriate concentrations is possible, but supplementing culture medium with serum from pregnant women provides a comprehensive approach.

Depending on the research focus, integrating multiple aspects is ideal to best represent pregnancy in vitro. Both two-dimensional and three-dimensional cultures can accommodate these factors, but cocultures or engineered tissues mimicking the heart and vasculature currently offer the most accurate representation (see Fig. 3).

Figure 3.

Figure 3.

Models to mimic aspects of pregnancy in vitro. Stem cells can be differentiated into cardiomyocytes and other cell types that can be studied in homogeneous cultures or in coculture systems (e.g., tissue engineering). Techniques for heart rate modulation, blood pressure modeling, and culture media adjustments are also depicted, highlighting the comprehensive approach to simulate pregnancy conditions. PBMC, peripheral blood mononuclear cell.

RELEVANT MODELS OF COMMON PREGNANCY-ASSOCIATED DISEASES AND RISK FACTORS

Hypertensive Disorders of Pregnancy

Hypertensive disorders of pregnancy (HDP) are leading causes of maternal and fetal mortality and morbidity, affecting 116 per 100,000 women of reproductive age globally (118). The most prevalent of these disorders are preeclampsia and related complications [i.e., eclampsia and hemolysis, elevated liver enzymes, and low platelet count (HELLP) syndrome], gestational hypertension, chronic hypertension, and preeclampsia superimposed on chronic hypertension (119121).

Preventative and therapeutic interventions to treat HDP and its cardiovascular sequelae are limited; thus, there is an urgent need for research on the mechanisms of HDP and therapeutics. Because of ethical aspects and restrictions in pregnant persons’ inclusion in clinical trials, the scientific community has turned their efforts to the development and use of experimental animal models. In vivo models range from large animals such as nonhuman primates, baboons, rhesus monkeys, dogs, and sheep to small animals such as rats, guinea pigs, and mice (122). Induction of HDP in an animal model is experimentally accomplished by disruption of maternal cardiovascular adaptations and pregnancy homeostasis. The most used experimental approaches include 1) surgical reduction of uteroplacental blood flow or placental ischemia (123133), 2) in vivo knock out or overexpression of genes using transgenic animals (82, 134145), 3) exogenous administration of pharmacological agonists or antagonists of key molecular pathways (146162), and 4) environmental stressors such as prenatal high-cholesterol diet (163168) or hypoxia (7) (see Table 2). Rodents with preexisting hypertension or other preexisting comorbidities superimposed by signs of preeclampsia during pregnancy have also been used to understand the early development of the syndrome (see Table 2) (169175).

Table 2.

Common pregnancy-associated diseases and relevant surgical, diet-induced, pharmacologically induced, genetically induced, and spontaneously induced animal models

Pregnancy-Associated Disease or Risk Factor Approach Model Species Hemodynamics Maternal Vascular Function Maternal Cardiac Function Postpartum Cardiovascular Complications Key Selected References (PMID)
Preeclampsia Surgical Uterine artery ligation Primates ↑BP ? ? ? 17377512
RUPP
(uterine and abdominal aortic occlusion)
Rats
Mice
↑BP
↑Uterine artery resistance
Endothelial dysfunction
Glomerular endotheliosis
↑ Vascular oxidative stress
↓ LV EF
Cardiac hypertrophy
Cerebral edema
Neuroinflammation
↓ LV EF
↓ Stroke volume
BP ↑ or normalized
Vascular dysfunction
25298513
10642326
31669926
29588233
28202440
34727994
27658290
31266978
Selective (s)RUPP (ovarian and uterine occlusion) Rats ↑BP
↑Uterine artery resistance
↑vascular oxidative stress ? ? 31266978
Diet induced High-cholesterol diet Rats ↑BP Endothelial dysfunction
Cerebrovascular dysfunction
? Cerebrovascular dysfunction 32856035
36689585
35137612
35615682
Genetic/Transgenic Angiotensinogen/renin Rats
Mice
↑BP Vascular dysfunction
Glomerular endotheliosis
↑Cardiomyocyte size ↑LV Mass
↓Global longitudinal strain
↑Cardiomyocyte size
↑Cardiac output
26394667
31786987
27927648
STOX1 Mice ↑BP ? Cardiac hypertrophy BP and
albuminuria
normalized
23357179
26758611
C1q−/− Mice ↑BP
↑stroke volume (postpartum)
↑end diastolic volume (postpartum)
↑end systolic volume (postpartum)
Vascular dysfunction ? ↓BP after delivery
↑BP late postpartum
Mild vascular dysfunction,
↑LV mass
↑myocardial cell hypertrophy and fibrosis
32374620
29669946
COMT−/− Mice ↑BP ↑Vascular dysfunction ? ? 22392899
18469803
Pharmacological/Infusion AVP Rats
Mice
↑BP Glomerular endotheliosis ? ? 30282823
25001273
TLR agonists Rats
Mice
↑BP Vascular dysfunction
↑Vascular oxidative stress
? ? 29437897
26873968
37352412
25853857
19779466
TNF-α Rats ↑BP Impaired
BBB
? ? 15928030
26400187
sFlt-1 Rats
Mice
↑BP Vascular dysfunction ? No long-term effects on BP and vascular function 17403433
19915174
20026766
20567176
Nω-nitro-l-arginine methyl ester Rats
Mice
↑BP Vascular dysfunction ? Hypertension resolved
Impaired vascular reactivity
36260752
25240245
Chronic AT1-AA Rats ↑BP Renal vascular dysfunction ? ↑ LV Mass
↑ Cardiac collagen deposition
Myocardial hypertrophy
21600854
24979132
Spontaneous Dahl Salt-sensitive (SS) Rats ↑BP
↑uterine artery resistance
↑Cerebral blood flow autoregulation
↑Myogenic tone
↑Blood brain permeability
? ? 25904684
30612494
33275518
BPH/5 Mice ↑BP
↑Uterine artery resistance
Decidual artery remodeling BP recovery after delivery 11882569
16957025
Diabetes Diabetic pregnancy Diabetic-NOD Mice ↓Mean uterine artery velocity
↓MAP mid to late pregnancy
Endothelial dysfunction Early LV dilation
No ↑ in LV CO
? 17827401
22014504
23636813
Gestational diabetes Db/+ Mice ↑Uterine artery resistance Endothelial dysfunction ? ↑Long-term endothelial dysfunction 21266665
20554929
Peripartum Cardiomyopathy Genetic/transgenic Cardiomyocyte STAT3−/−
and
Cardiomyocyte STAT3−/− with caAKT
Mice No change in HR Increased Vasohibin,
Endothelial apoptosis and dysfunction,
Vascular insufficiency
Capillary dropout
Systolic heart failure (after 1–2 pregnancies)
↓ FS%
↑ HW
↑ LVEDD, LVESV
Energetic deficits
fibrosis
Systolic heart failure and cardiomyopathy (after 1–2 pregnancies)
17289576
28201733
24448315
Cardiomyocyte PGC1-α−/− Mice ↓HR ↓endothelial gene expression
Capillary dropout,
↓microvascular density,
↓VEGF
Systolic heart failure (after 1–2 pregnancies)
↓FS%
↑HW:TL
↑LVEDD, LVESD,
E/E
Dilated cardiomyopathy (after 1–2 pregnancies) 22596155
Titin (TTN) mutation/ TTNtv knockin Mice ? ? TTNtv mutation similar that seen in women with PPCM,
↑fibrosis
↑LVEDD
↓ FS%
? 24558114
19406126
Cardiomyocyte Ndufs4−/− Mice ? ? ↑HW:TL
↓FS%
↑LV dimension
Cardiomyopathy (after 1–6 pregnancies) 23931755
Cardiomyocyte Perk−/− Mice No change in BP Capillary dropout, ↓VEGFa Systolic heart failure (after 1–2 pregnancies)
↓FS%
↑HW
↑LVEDD,
Oxidative stress and protein aggregates,
apoptosis
Cardiomyopathy and ↑mortality (after 1–3 pregnancies) 34548576
Cardiac NPR1−/− Mice No change CO ? ↓FS, EF,
↑LVEDV, LVESV,
↑fibrosis, myocyte size, HW:TL (during lactation period).
Cardiomyopathy and ↑mortality, fibrosis, myocyte size, HW:TL (after 1–5 pregnancies) 31665900
Pharmacological Notch1 inhibition (with LY-411575) Mice ? Capillary drop out,
↓VEGFa
↓FS, EF
↑LVEDD, LVEDV, LVESD, LVESV
↑HW:TL and myocyte size
↑fetal gene expression
fibrosis
↓FS, EF
↑LVEDD, LVEDV, LVESD, LVESV
↑HW:TL and myocyte size
↑fetal gene expression
↑fibrosis
32529705
ErbB1/ErbB2 inhibitor (Lapatinib) Rats and mice No change in HR ? ↑Mortality,
↑HW:TL
↓ FS, EF
↑LVEDD,
No fibrosis
No apoptosis
Cardiomyopathy 21186272
Environmental exposures Nano-TIO2 (Dose range: 9–12 mg/m3 for 4–6 h/day; number of maternal exposure days varies within studies). Rat ↑Uterine artery vasoconstriction
↓Placental blood flow
Uterine artery endothelial dysfunction
Impaired placental vascular reactivity
↓Mitral value deceleration time
E/A ratio
↓Female pup weight
↑Oxidative stress in Dam and Offspring
35642938
35066857
36950144
35260159
37181648
30734150
31215478
Ozone
(Dose: 1 ppm O3 for 4 h on GD10.5)
Mice ? ? ↓SV
↓LV EF
Altered transcriptomics
? 37695302
Ozone
(Dose: 0.4–0.8 ppm O3 for 4 h/day on GD5 and 6)
Rat ↑Uterine artery resistance ? ? ? 29269335
Diesel Exhaust
(Dose: 0.3 mg/m3 for 6 h/day, 5 d/wk, from GD0.5 to 17.5).
Mice ? ↑Increase embryo reabsorption
↑Placental hemorrhage
? 12-wk-old offspring: increased susceptibility to pressure-overload-induced heart failure 24533117
Electronic cigarette (Dose: aerosol mass or chemical concentration not reported – exposure condition was 3 h/d, 5 day/wk, from GD520). Rat ↓Uterine artery and fetal blood flow ? ? ↓Offspring weight
↓Crown-rump length
30653941

BBB, Blood brain barrier; BP, blood pressure; CO, cardiac output; EF, ejection fraction; FS, fractional shortening; GD, gestational day; GFR, glomerular filtration rate; HW, heart weight; LV, left ventricular; LVEDD, LV end-diastolic dimension; LVEDV, LV end-diastolic volume; LVESD, LV end-systolic dimension; LVESV, LV end-systolic volume; MAP, mean arterial pressure; PGC-1α, peroxisome proliferator-activated receptor-γ coactivator 1-α; PPCM, peripartum cardiomyopathy; RUPP, reduced uterine perfusion pressure; TL, tibia length; TLR, Toll-like receptor.

When deciding which HDP animal model is appropriate to address a research question, the following concepts should be considered:

  • 1) 

    Comparative physiological variation between humans and the experimental species of interest (176). In addition to variations in placental morphology and function (77, 177), the maternal cardiovascular adaptations and the maternal immune and hormonal changes during pregnancy and postpartum should be contemplated in a comparative fashion.

  • 2) 

    Heterogeneity of clinical presentation and potential clinical subtypes of HDP (178). For example, although an animal model may not recapitulate the heterogeneity in the diagnostic onset of preeclampsia (i.e., early vs. late-onset preeclampsia), it is still valuable in studying other cardiovascular features of the syndrome, such as maternal endothelial dysfunction, increased maternal blood pressure, impaired maternal cerebrovascular function, or maternal cardiac systolic and diastolic function.

  • 3) 

    Opportunity to study later-life maternal and offspring health. Animal models provide preclinical evidence for mechanistic pathways to later-life cardiovascular health. Because of shorter lengths of gestation and life spans, experimental models afford feasibility for long-term follow-ups of the mother and the offspring after a pregnancy with HDP.

  • 4) 

    Other considerations: gestational age and duration, timing of treatment or induction of HDP, time points of postpartum assessment for early versus late postpartum, and the relation of these reproductive stages to animal’s life span and expected reproductive transitions.

Gestational Diabetes and Diabetic Pregnancies

Pregestational and gestational diabetes are both characterized by elevated blood glucose, which can lead to both maternal and fetal complications (179). The incidence of women with preexisting diabetes is ∼2%, and the global incidence of gestational diabetes is 14% (179, 180). Women with gestational diabetes exhibit subclinical left ventricular structural changes, and those with insulin-dependent diabetic pregnancy display early restrictive filling patterns (181, 182). In addition, pregnant diabetic subjects exhibit endothelial dysfunction and altered uterine-placental vasculature (183, 184). To study the impact of maternal diabetes, animal models are frequently used (185). Animal models of diabetic pregnancy have hyperglycemia at the onset of or starting in early pregnancy, whereas models of gestational diabetes should have hyperglycemia onset later in pregnancy that ideally reverses with parturition (186). Blood glucose levels vary with food intake, thus fasting glucose levels are routinely compared. Blood glucose concentrations are monitored with a glucometer validated for the species or by biochemical assay (187). For most rodents, fasting blood glucose approximates 100 mg/dL. In rodents, mild hyperglycemia is ∼120–200 mg/dL and severe hyperglycemia over 250 mg/dL, although the cutoffs for mild and severe are subjective (185, 188).

Diabetic animal models include surgical pancreatectomy and viral approaches, but chemically induced and genetic models are the most common (see Table 2) (84, 186, 189192). Chemical treatment with streptozotocin (STZ) or alloxan damages β cells, and animal age, dose, and timing relative to mating will determine the degree of hyperglycemia (193). The effects of STZ and alloxan are mostly irreversible making their use a more appropriate model of diabetic pregnancy rather than gestational diabetes. Using an insulin receptor antagonist to block insulin action in pregnant mice is reversible, making this model more representative of gestational diabetes (194).

Genetic rodent models of diabetic pregnancy include nonobese diabetic (NOD), Ins2 Akita, New Zealand Obese mice, specific strains of C57BL/6 mice, and Zucker diabetic fatty, GK, BB, and Tet29 rats (185, 186). NOD mice undergo spontaneous β cell destruction between 12 and 30 wk of age resulting in severe hyperglycemia (195). Because NOD mice become diabetic at different ages, this model has the advantage of providing age-matched nondiabetic controls. Heterozygous Ins2 Akita possess a mutated Ins2 gene and are a diabetic pregnancy model with a range of glucose levels allowing evaluation of both mild and severe maternal hyperglycemia (196).

Few rodent genetic models mimic human gestational diabetes. Db/+ mice, heterozygous for a leptin receptor mutation, do not initially become diabetic. They develop moderate glucose intolerance during pregnancy that resolves after parturition, making them a good model for gestational diabetes (185). A less well-characterized gestational diabetic model, C57BL/6N mice, also acquires glucose intolerance during pregnancy (186).

Larger animal models such as nonhuman primates, sheep, pigs, and dogs have also been used as with surgically and chemically induced diabetes; however, high cost and ethical concerns limit studies (193) (see Table 1). Other models of diabetes during pregnancy, such as C57BL/6J mice, can be achieved through dietary manipulation, such as high-fat diet (186).

Myocardial Ischemia in Pregnancy

The drastic changes in the cardiovascular system during pregnancy are usually well tolerated in healthy individuals. However, some healthy individuals can develop severe adverse cardiac events in late pregnancy (197) such as myocardial infarction (MI) (198, 199) which has increased recently because of rising maternal age, smoking, chronic hypertension, obesity, and diabetes (200203). Pregnancy-associated MI accounts for over 20% of maternal cardiac deaths (199). The relative risk of MI during late pregnancy is approximately three- to fourfold higher than the rates in age-matched nonpregnant individuals (203). MI during late pregnancy and the peripartum period carries a markedly worse prognosis than in nonpregnant people for unclear reasons (202, 204). Eghbali’s (205, 206) laboratory successfully modeled this observation in rodents using both in vivo and ex vivo models of ischemia-reperfusion injury. In an in vivo model, nonpregnant and late-pregnant rats underwent occlusion of the left anterior descending coronary artery for 45 min, followed by 3 h reperfusion (205). At the end of the reperfusion, Evans blue dye was injected to assure both experimental groups were exposed to the same ischemic risk. This in vivo model showed myocardial infarct size in late pregnant animals was approximately fourfold greater than in nonpregnant animals (205). To examine the translational potential of gene therapy or microRNA delivery, it is necessary to extend the reperfusion duration to 24 h, allowing sufficient time for regulation of the expression of target genes.

In the ex vivo Langendorff perfusion, isolated mouse hearts were subjected to a 20-min ischemic event followed by 40 min of reperfusion (205, 206). This model allows for the real-time measurement of hemodynamic parameters at the baseline, during ischemia, and at the reperfusion. Although, the hemodynamic parameters were similar between nonpregnant and late pregnant before ischemia as assessed by the rate pressure product, an indicator of the oxygen requirements of the heart and measured as the left ventricular developed pressure × heart rate, the functional recovery of the heart was extremely poor in late pregnant mice upon ischemia compared with nonpregnant mice (205). Remarkably, there was partial recovery in all hemodynamic parameters observed in hearts in mice 1 day postpartum, and even more improvements were noted in hearts 7 days postpartum (205). As the first few minutes of reperfusion is very critical for any intervention, this ex vivo model is also very effective for measurements of calcium retention capacity and signaling pathway analysis, in which the duration of reperfusion should be reduced to 5–10 min (205, 206).

Hypoxia Models

Gestational hypoxia arising from anemia, smoking, sleep apnea, and residence at high altitude (HA > 8,250 ft) plays an important role in the etiology of pregnancy complications, including fetal growth restriction (FGR) and preeclampsia (207, 208), and exerts long-term impacts on offspring health (76, 209). Pregnant women living at HA are exposed to chronic hypobaric hypoxia. In Leadville, Colorado (3,100 ft), for instance, the partial pressure of inspired oxygen is ∼33% lower than at sea level (100 mmHg vs. 150 mmHg) (210). HA pregnancy therefore provides a natural in vivo model for understanding the mechanisms by which hypoxia influences placental function and hypoxia-related disorders of pregnancy. HA reduces birth weight (211213), increases the incidence of FGR, and hypertensive disorders of pregnancy (214216) as well as maternal susceptibility to cardiovascular dysfunction during and long after pregnancy (217). It has also been shown to reduce the normal rise in uterine artery blood flow (216, 218, 219) and cardiac output (220), and blunt myometrial artery relaxation with exposure to specific vasodilatory agents (221, 222); these effects are reminiscent of the impaired vasodilation and remodeling of uterine vessels, lesser uterine artery blood flow, and placental insufficiency often observed in preeclampsia and FGR (223227). Placental morphology, function, and transcriptional profiles have also been reported to be affected by HA (228231). Advantages of the human model include the ability to study the effects of hypoxia without the confounding effect of underlying disease and the enhanced potential for translational impact. Differences between ancestry groups with respect to the magnitude by which high altitude reduces fetal growth (212, 216, 232) are also valuable for determining the role of genetics for hypoxia-related disorders of pregnancy. One limitation is the inability to serially sample the placenta and umbilical or uterine vessels across gestation. This prohibits access to the samples required for directly establishing the impact of gestational hypoxia on transplacental oxygenation or nutrient exchange during pregnancy and determining whether such effects precede or follow the onset of fetal growth restriction or maternal vascular dysfunction.

Various animal models have been developed to understand the pathophysiology of human pregnancy complications associated with hypoxia and to explore the potential intervention of these disorders. Commonly used animal models of gestational hypoxia include rodents and sheep, achieved by exposing the animals to normobaric hypoxia ranging from 10% to 14% O2 at approximately sea level or to hypobaric hypoxia at high altitude during different states of gestation (7, 62, 76, 233240). The chick embryo is also frequently used to study impacts of chronic hypoxia on fetal growth and the developing cardiovascular system independent of maternal and placental effects (241). These models have successfully recapitulated many features of preeclampsia and fetal growth restriction and demonstrated desirable efficacy of trial drugs (7, 79, 235, 242, 243). To more precisely mimic human conditions, factors such as placenta type, gestational duration, and metabolism rate should be taken into consideration when selecting an animal model. For example, the guinea pig is preferred if placentation is studied as it shares with humans the hemochorial placental type (77, 244). Knockout (KO) mice are useful tools to link the function of a gene and its pathway to the pathophysiology of pregnancy complications associated with gestational hypoxia (144, 145). Moreover, the sheep model is the choice if fetal instrumentation and physiology recording are required (76, 207).

Peripartum Cardiomyopathy

Peripartum cardiomyopathy (PPCM) is an idiopathic systolic dysfunction that occurs in the last trimester of pregnancy and during the first 6 to 12 mo postpartum. Although rare, the incidence of PPCM is increasing, currently impacting around 1:1,000 to 1:3,000 births, each year in the United States (245). Several risk factors and comorbidities contribute to the development of PPCM, such as advanced maternal age, high blood pressure, multiple gestations, and lactation (245). Significant racial disparities exist, with black and African-American women being disproportionately impacted by PPCM.

The etiology of PPCM is poorly understood. Currently, mutations in ∼21 genes have been linked with PPCM, with 15% of PPCM arising from mutations in several sarcomeric proteins, including titin, myosin heavy chain 6, and myosin heavy chain 7 (176). However, it is important to note that mouse models harboring these mutations have either not been examined in the context of pregnancy or do not recapitulate the PPCM phenotype in mice. This also means that around 85% of PPCM originates from nongenetic causes. Some studies suggest increased risk of PPCM following viral infection and exaggerated immune responses. However, much of what has been uncovered regarding the underlying mechanisms contributing to PPCM indicates key roles for hormonal changes (i.e., prolactin and its cleavage) and angiogenic imbalance (i.e., antiangiogenic form of prolactin and increased circulating levels of sFlt1) (246, 247).

Around 12 mouse models exist that recapitulate many of the key characteristics of PPCM (176) (see Table 2) (246256). Of these mouse models, the cardiomyocyte-specific signal transducer and activator of transcription 3 (STAT3) (251) and cardiomyocyte-specific peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) KO mice (247) are among the most widely used. These models recapitulate the hormonal and angiogenic imbalance seen in women with PPCM and can be rescued using the prolactin secretion inhibitor, bromocriptine. However, despite their use, both STAT3 and PGC1α have not been shown to change in the hearts of patients with PPCM. In addition, the impact of comorbidities and risk factors known to contribute to PPCM in women, such as aging, multiple gestations, and so forth have not been previously considered in these models. At present, none of the existing models fully recapitulate the PPCM phenotype so caution must be taken when selecting a model for use. Moving forward, additional models are required to further clarify underlying mechanisms contributing to PPCM.

Advanced Maternal Age

Defining advanced maternal age and its impact on pregnancy.

Advanced maternal age pertains to pregnancy in women and females aged 35 yr or older and is associated with an increased risk of complications such as preeclampsia, intrauterine growth restriction, preterm birth, stillbirth, and gestational diabetes mellitus (257261). Understanding the drivers of increased complications is crucial due to delayed childbearing becoming increasingly common place, necessitating effective clinical management and intervention opportunities. Moreover, there is emerging concern about the long-term cardiovascular health of postpartum mothers and offspring born to older mothers (257, 262265).

Impact of aging on cardiovascular adaptations.

Increased susceptibility to pregnancy-related complications in women of advanced maternal age may, in part, stem from inadequate cardiovascular adaptations during pregnancy. Aging effects encompass reduced vascular compliance, impaired endothelium-dependent function, and heightened activity of the sympathetic nervous system, contributing to increased arterial stiffness and hypertension (257, 266, 267). Evidence from rodent models and in women indicates that advanced maternal age impacts vascular function in pregnancy (257, 268271).

Methodological recommendations for studying cardiovascular function at advanced maternal age.

For investigating cardiovascular adaptations in advanced maternal age, consider the age of animal models (and the impact that will have on fertility) and age-associated body weight changes. In both human and animal studies, select the appropriate vascular bed for investigation (268273).

To measure cardiovascular function in advanced maternal age, a variety of commonly used techniques can be used:

  • 1) 

    Doppler ultrasound: a noninvasive imaging technique to assess blood flow velocity in maternal and fetal vessels (257). This requires the use of anesthesia (most commonly isoflurane) that can be considered a confounding factor. However, both control and experimental animals should be treated the same way, and care should be used to minimize the impact by paying close attention the animal’s body temperature (often a small space heater is required in addition to a heated animal handling table), heart rate, and breathing rate, and adjusting the anesthesia as required (it is therefore advantageous to use an inhaled anesthetic such as isoflurane, where adjustments can be made to the level of anesthesia).

  • 2) 

    Maternal blood pressure measurements: performed noninvasively using a sphygmomanometer or continuously through telemetry devices in animal studies (7, 257, 268, 271, 274). Telemetry devices are the gold standard because they do not require the animal to be restrained, but their use in female mice (especially young mice) can be challenging because of the size of the telemetric transmitter that needs to be implanted; a person highly skilled in such small animal surgery will have greater success. Ideally, surgery should be performed before pregnancy to prevent additional impacts on the pregnancy. Using a sphygmomanometer is quite simple but can cause stress to the animals since they are restrained in acrylic holders, which can add variability to the data, and could impact the pregnancy outcomes. An appropriately sized restraint should be selected, taking into account that the animal might be pregnant, and a larger restraint might be required in later pregnancy. Intra-arterial catheters can also be used for blood pressure measurements in both conscious and anesthetized animals.

  • 3) 

    Isolated vascular studies: In animal models and tissue from human pregnancy, small blood vessels can be isolated to study their contractile and relaxation responses, as well as arterial stiffness using wire myographs and pressure arteriographs. Using pressure arteriographs, ex vivo vascular compliance, myogenic tone, and flow-mediated vasodilation can also be assessed. One potential confounding variable in pregnancy research is the lack of capacity to hold preconstricted vascular tone in some vascular beds to assess vasorelaxation. In addition, for the pressure arteriography, selection of vessels without branches is essential.

  • 4) 
    Cardiac function:
    • 1) 
      echocardiography offers insights into cardiac structure and function and has been measured in offspring of advanced maternal age dams (263) and can also be measured in the dam. Anesthesia and body temperature, heart and respiratory rate considerations outlined in the Doppler ultrasound section are also relevant for echocardiography.
    • 2) 
      Continuous, noninvasive measurements of cardiac output can be estimated using the bioreactance method. This method involves the provision of alternating high-frequency electrical current that traverses the thoracic cavity, in which four surface electrodes collect information on relative phase shifts of current, which are used to calculate blood flow (see Ref. 275 for additional information on the bioreactance method). This method can also be used to collect information on stroke volume, heart rate, peripheral vascular resistance, and mean arterial pressure (274).
    • 3) 
      Isolated working heart studies: Assess capacity to recover from ischemia-reperfusion injury (257) and offers the ability to study basal as well as simulated cardiac function. Preliminary experiments may need to be performed to determine the optimal period for ischemia, especially in older animals. A previous study in offspring from advanced maternal age dams reduced the global ischemia period to 10 min as none of the hearts were recovering with the standard 20 min ischemia (262).

Understanding the impact of advanced maternal age on pregnancy outcomes, including cardiovascular adaptations, warrants further exploration. Advanced cardiovascular measurement techniques may enhance the understanding and optimize care for older expectant mothers.

CONSIDERATIONS FOR RIGOROUS EXPERIMENTAL DESIGN

Timed Pregnancy Studies

Timed pregnancy studies afford investigators the opportunity to directly examine time points during pregnancy (see next section for time-point selection), which is particularly advantageous if a period of vulnerability has been identified for examination of physiological experimental endpoints or specific timed inventions. For timed murine pregnancy studies, estrus should first be stimulated in female mice using the concepts put forward by Whitten (276), whereby soiled bedding containing male pheromones is placed into the female cage to stimulate estrus. Because the female estrus cycle in rodents is 4–5 days (277), male studs should be paired with female mice until either the presence of a copulatory plug or if time-points postpartum are of greater interest, until one cycle has elapsed. Male mice should be paired with females in the early evening and removed and single housed the next morning and plugs checked early and daily because sometimes these can fall out or dissolve. Once a copulatory plug is identified this signifies gestational day 0.5. A vaginal swab to confirm the presence of sperm in the vagina can be performed as per Caligioni’s (277) and Byers et al.’s (278) study, as an additional confirmation of copulation; however, it is important to note that this is somewhat invasive and can result in stress. Because the presence of a copulatory plug and the use of vaginal swabs only indicates copulation has occurred rather than confirming pregnancy, regular body weight measurements can be obtained as a secondary confirmation of pregnancy. This is advantageous in strains with characteristic weight gain during pregnancy, such as FVB/NJ mice, which gain ∼3–4 g in the first 8 days following the identification of a copulatory plug (279). Although ultrasound is a commonly used methodology to assess the human pregnancy stage and has been used to determine the pregnancy stage in rodents (280), this is less commonly used in the field for pregnancy determination in animal models.

Parity, Gravidity, and Time Points

The use of preclinical animal models to investigate physiological and pathophysiological adaptations during pregnancy and postpartum is well accepted. Yet, several factors should be considered when designing preclinical models for studying healthy pregnancies, pregnancies with complications such as HDP, and postpartum physiology. Key considerations include timing of interventions and end point measurements. With studies focused on the assessment of cardiovascular physiology during an uncomplicated pregnancy, selecting time points for investigation is often impacted by the type of animal and the strain chosen. For example, the commonly used mouse strains, FVB/NJ and C57BL/6, although both mouse strains have different gestational lengths (281) so care must be taken when selecting time points. In addition, time-point selection will likely also be governed by the primary end point of the study and the time frame during pregnancy that it is likely to be changed. If the latter is unknown, investigators should select time points across pregnancy and postpartum.

Key considerations should also be made for studies focused on pregnancy-associated diseases. For example, many experimental models of HDP, such as preeclampsia, include surgical (282), pharmacological (54, 165, 283, 284), dietary (165), environmental (7), or immunological (285287) approaches (Table 2) that are initiated in mid to late gestation, mimicking midgestational manifestation and diagnosis of preeclampsia. Genetic approaches to replicate HDP begin before pregnancy (134, 288) allowing for investigation of early gestational changes before the development of the hypertensive phenotype (134) (Table 2). Several preclinical models develop hypertension during pregnancy mimicking superimposed preeclampsia (169, 289, 290) where preeclampsia symptoms arise in the presence of preexisting hypertension. Thus, many experimental models are limited to study of late gestational events whereas others allow for study of mechanisms related to early gestational events but often in the presence of superimposed preeclampsia (169, 289, 290). Exceptions include the rodent model of preeclampsia induced via gestational infusion of arginine vasopressin that is associated with alterations in placental morphology in mid gestation (158) and the rodent model of preeclampsia achieved through midgestational treatment with leptin (54).

In rodents, multiparity can influence maternal body composition and inflammation, contributory mechanisms in the etiology of preeclampsia (291). In terms of long-term postpartum health, clinical studies report that multiparity is associated with decreased cardiovascular health scores (292), decreased hippocampal volumes, and worse memory scores in women with five or more deliveries (293). In rodent models of stroke, multiparity results in smaller infarcts and reduced neuroinflammation despite having features of increased metabolic risks (294). It is also challenging to separate multiparity from aging. Advanced maternal age is associated with adapted responses resulting in beneficial effects on maternal blood pressure and vascular function in late gestation in dams that deliver pups at term (268), which is unique to rodents. Vascular function is worsened postpartum in dams with advanced age, in particular those that do not deliver (264). Thus, use of first-time pregnant rodents may reduce biological variability for studies with a focus on pregnancy and postpartum outcomes (165). An additional caveat to the use of preclinical models involves placental structure that can differ based on species, human compared with rat (295) (See Table 1). However, rodents, which have a gestational period of ∼3 wk, are extensively studied and numerous investigators report that maternal renal and systemic hemodynamic changes in the rat mimic the time line in the human (296298) highlighting that benefit for rodent models.

Pharmacological Properties of Drugs during Pregnancy

Many preexisting conditions demand continuation of maternal pharmacological treatments throughout pregnancy and postpartum. Mothers must also be treated for diseases they acquire during pregnancy or postpartum and maternal vaccination may occur during the perinatal period. In addition, pregnancy-induced complications require the use of maternal therapeutics. Experimental animal models provide a platform for testing drug efficacy, side effects, and mechanisms of action that can aid in safe and efficacious implementation of an intervention.

Pregnancy and the postpartum period are distinct physiological states, involving multisystem adaptations that significantly affect pharmacological properties of drugs (299, 300). Little is known about the interaction between the early postpartum period and drug properties. Furthermore, there is a paucity of research on the effects of maternal therapeutics during pregnancy on long-term maternal health outcomes. The implementation of a maternal treatment during pregnancy or postpartum is a dilemma because while a treatment may reduce maternal symptoms, it can impair offspring development via placental transfer during gestation or milk transfer during lactation (301). On the other hand, this exact same principle of maternal-fetal communication is the basis of the systemic maternal treatments to improve fetal outcomes.

Key considerations for drug properties and their efficacy on pregnancy-associated physiology.

  • 1) 

    Increase in the volume of distribution of hydrophilic substrates (302305). Impact: dosing of hydrophilic drugs.

  • 2) 

    Serum albumin is reduced, and compensatory respiratory alkalosis develops. Impact: Drug bioactivity and changes in blood acid/base influence breathing patterns, which can impact exposure conditions for inhalation studies (see Environmental Exposures).

  • 3) 

    Increase in glomerular filtration rate. Impact: elimination rates of drugs metabolized by the kidneys and their half-life.

  • 4) 

    Gastric mobility is reduced during pregnancy. Impact: bioavailability of orally administered medication and/or alter time for ingestion-related exposures (see Environmental Exposures).

  • 5) 

    Drug metabolizing enzymes change during pregnancy and sex steroid concentrations are increased. Impact: drug metabolism.

  • 6) 

    Nausea and vomiting are common features during pregnancy. Impact: drug absorption may be reduced. This may not be an issue in preclinical rodent models in initial testing of drug efficacy they lack the emetic reflex (306).

Lactation and Breastfeeding

Lactation and breastfeeding are associated with maternal cardiovascular health benefits (307). Lactation is the period that starts right after delivery and ends once the mother’s body has ceased to produce milk. Breastfeeding or nursing is when a mother feeds her baby using breasts. Infants could also be fed milk that mothers have expressed or pumped and saved in a bottle. Thus, because of the different ways of breastfeeding, the infant defines maternal physiology during this period.

The duration of lactation differs among the species and is dependent on the maturity of the baby at birth. In precocial species, newborns are considerably independent, their behavior and morphology are more mature, and they can survive with limited support (308). Babies unable to move independently and requiring extensive parental care and food provision are born to altricial species (308). Rodents, extensively used in pregnancy and lactation studies, are altricial species: their newborns are ready for life by themselves at 3 wk of age. Thus, rodent dams’ lactate for 3 wk (309). Sheep and nonhuman primates are precocial species. Artificially, sheep are weaned at 1–2 days of age. However, wild sheep can nurse for up to 12 mo (310). Lactation in nonhuman primates lasts 71–551 days, depending on the species. Nonhuman primates that undergo a menstrual cycle experience a prolonged lactation-induced anovulatory period: lactation amenorrhea, which needs to be taken into consideration when designing experimental studies (311). Finally, humans are the altricial species since the newborns are helpless at birth and require much parental care (312). In humans, the American Academy of Pediatrics and the World Health Organization recommend that newborns be exclusively breastfed until 6 mo of age (313).

Several experimental conditions are specific only to the lactation period, significantly impacting the study outcomes. Study groups in women should be defined according to lactation and breastfeeding status: exclusive breastfeeding, mechanical breast pumping, bottle feeding, and any combination of the aforementioned.

Several hormones are released only during lactation and breastfeeding and are known to affect the maternal cardiovascular system. Among them, oxytocin is the most studied. In addition to its role in parturition, lactation, and modulating social behavior, oxytocin lowers blood pressure, heart rate, and obesity (314). Moreover, breastfeeding mothers have higher oxytocin levels in plasma than formula feeders, and it was associated with lower blood pressure and heart rate. Breastfeeding induces immediate and short-lasting (20 min) oxytocin release (315). Oxytocin is released in pulses that become longer as lactation proceeds (315). Thus, experiments should be timed accordingly and under the same conditions for each mother: during breastfeeding, 10 min (as an example) before and after breastfeeding.

Other confounders or study group determinants for studies in women are mode of delivery (spontaneous or C-section); duration of pregnancy (term or preterm); pregnancy complications (gestational diabetes, preeclampsia); nutrition; smoking status; prior history of breastfeeding; psychosocial stress; and medications. All these conditions influence lactation, breastfeeding, and oxytocin production and release, subsequently influencing the cardiovascular system (316). For animal studies, one must remember that if the mother is taken away from her pups for any period, there is always a risk that the dam might not recognize her offspring and refuse to nurse them leading to neonatal mortality (317).

ACCOUNTING FOR ENVIRONMENTAL FACTORS AND MODIFIABLE STRESSORS

Diet and Obesity

Diet choice.

There are several key considerations to make when establishing models of dietary manipulation during pregnancy. In terms of modeling Western-style diets, there are two main approaches 1) using high-fat diets (318) and 2) using high-fat high-simple carbohydrate obesogenic diets (319). Rodents are generally good at regulating food intake in response to a high-fat diet, and therefore often do not gain excess weight when fed ad libitum such a diet (unless the feeding regimen is over an extended period). In contrast, diets rich in simple carbohydrates generally override natural satiety signals. Therefore, feeding of such diets ad libitum leads to increased food intake, weight gain, and increased adiposity and other aspects of metabolic dysfunction.

Standardization of maternal physiology.

There may be interanimal variation in response to diet, for example in nonhuman primates it has been reported that in response to a high-fat diet, females can be divided into responders (i.e., gain excess weight) or nonresponders (i.e., do not gain excess weight) (320). It is therefore important to record maternal weight (and ideally body composition) longitudinally. In addition, longitudinal maternal blood sampling to allow measurement of hormones such as insulin and glucocorticoids and metabolites such as glucose is advisable. This will allow correlation analysis between maternal physiology and cardiovascular phenotyping and ultimately if interventions are tested, allow causal relationships to be identified.

Minimizing external factors in offspring.

When addressing maternal effects on offspring, the mother represents the statistical unit. Therefore, when carrying out studies in a litter-bearing species such as mice, this can be capitalized on, and littermate siblings used to address the effect of offspring sex or diet (e.g., by weaning half of the litter on to a chow diet and the other half on an obesogenic diet). Feeding status (fed or fasting) should also be standardized as should the time of day when experiments are performed (noting that rodents are nocturnal so will feed during the dark phase). Offspring measurements should also be carried out longitudinally, with the timing of phenotyping (e.g., before/after sexual maturity, before effects of altered diet results in changes in body composition, etc.) also considered.

Fitness and Exercise Considerations in Human Pregnancy

Current guidelines recommend engaging in at least 150 min of moderate-intensity physical activity on three or more days of the week during pregnancy to derive clinically meaningful health benefits (321, 322). These include a ∼40% reduction in the risk of developing preeclampsia, gestational hypertension, gestational diabetes, and a reduced risk of depression by nearly 70% (323, 324). An individualized and gradual resumption of physical activity is encouraged following delivery, with appropriate rehabilitation of pelvic floor dysfunction, musculoskeletal pain, and other symptoms that may limit exercise to support physical and mental health (325, 326).

Exercise training is associated with cardiovascular health benefits during pregnancy including a small increase in fitness (↑V̇o2max ∼2.8 mL/kg/min) and reduced resting heart rate and blood pressure (327). Exercise training is anticipated to improve vascular health (e.g., endothelial function and arterial stiffness) during and following pregnancy, especially in those who have underlying cardiovascular dysfunction (328). Given the impact of physical activity and exercise on cardiovascular health, an objective (i.e., accelerometer) or subjective (i.e., questionnaire) assessment of physical activity is recommended in conjunction with cardiovascular assessments in pregnancy and postpartum.

Emerging data from women have identified an association between cardiorespiratory fitness and HDP (329). Before fitness testing, it is essential to screen for medical conditions where exercise is not advised (e.g., severe preeclampsia). The “Get Active Questionnaire for Pregnancy” was developed for this purpose (https://csep.ca/2021/05/27/get-active-questionnaire-for-pregnancy). Fitness testing to maximal intensities is reserved for experienced researchers who perform concurrent fetal monitoring. Submaximal exercise tests where the peak exercise intensity falls within the recommendations of current guidelines are considered to be low risk in those without contraindications (330), and fetal monitoring is optional.

Key considerations for exercise testing and training in pregnant human subjects.

  • 1) 

    Maintain adequate nutrition and hydration before, during, and after exercise.

  • 2) 

    A nutritious snack ∼1 h before exercise is recommended to maintain maternal glucose concentrations, especially before longer duration and or higher intensity activities.

  • 3) 

    Avoid exercise in excessive heat, especially with high humidity.

  • 4) 

    Complete a preexercise assessment of resting heart rate, blood pressure, and glucose so that these values can be interpreted within the context of pregnancy/postpartum physiological adaptations and identify undiagnosed abnormal values.

Detailed recommendations and considerations on exercise testing during pregnancy have been previously described by Wowdzia and Davenport (331). Guidelines for cardiorespiratory exercise testing specific to the postpartum period have not been developed. Adult testing guidelines (e.g., American College of Sports Medicine) may be used following an individualized and gradual resumption of moderate-to-vigorous intensity physical activity after childbirth. It is essential to be aware of how the unique considerations of the postpartum period (e.g., type of delivery and healing times, impact of sleep deprivation and breastfeeding, complications persisting from pregnancy or newly developed postpartum) may impact the results and or applicability of the test. Detailed recommendations on appropriate milestones and progression toward moderate-to-vigorous intensity physical activity are outlined by Christopher et al. (332).

Environmental Exposures

Environmental exposures generally fall into three categories, inhalation, ingested, and dermal (see Table 2) (333343). When designing any exposure experiment it is important to determine the media for the contaminate (e.g., air, liquid, or solid media) and route of exposure (e.g., inhalation, ingested, and dermal) that is most relevant to the environment and biological outcome under investigation. It is always critical to understand and report the “exposure dose.” Dosing related to liquid and solid media often seems more intuitive and more easily assessed with standard laboratory equipment, whereas dosing from inhalation exposures typically requires more specialized instruments and expertise and may be more complex depending on the desired measurement. It is important to emphasize that solely describing the exposure conditions (e.g., the number of hours of exposure) is not reporting the “dose” of the exposure. This error is most often made with inhalation exposures when reporting only the number of puffs or time in the chamber rather than reporting the size and/or concentration of the airborne media (i.e., particulate matter, PM). Depending on the experimental question or conditions, the chemical composition of PM (or gaseous phase) may also be relevant, and will require additional expertise and equipment to obtain, interpret, and determine biological relevance.

Regardless of media or route, there are different approaches that can be used when assessing dose (344). The potential dose, which is the amount of contaminant brought into or on the body, is commonly used for inhalation exposures, and in particular with pregnancy exposures when the objectives require study of progeny after birth. The “potential dose” gives a measure of the amount of contaminate or PM that is brought into the mouth/nose or on the skin. But it is important to realize that this measure does not account for what is absorbed.

The applied dose is the amount of contaminant entering the body at the absorption barrier (e.g., reaching the respiratory tract, gastrointestinal tract, or absorbed by the skin). Since most inhalation exposures (including during pregnancy) are attempting to mimic or replicate a deviant environmental condition (i.e., air pollution, smoking, occupational environment, etc.) reporting the “potential” or “applied dose” is the minimal standard to provide rigor and reproducibility. For inhalation exposures, the “applied dose” (e.g., lung burden) can be estimated from the “potential dose” using several validated mathematical modeling approaches, [such as, multiple path particle dosimetry (MPPD), computational fluid dynamics (CFD), etc.]. Interested readers are directed to the following references for more details on the modeling approaches (345347).

Finally, measuring the internal dose or biologically effective dose will typically provide better correlation and understanding of toxin-related exposures, but may not always possible or practical to obtain. The “internal dose” is the amount of contaminant that gets past the absorption boundary and into the blood, whereas the “biologically effective dose” is the amount of contaminant that interacts with the internal target tissue or organ. Whether this is possible/practical to measure will depend on the contaminate and/or study context. For example, the “internal dose” is often better suited to single toxin effects. In contrast, complex PM (e.g., cigarette smoke, e-cig aerosol, urban wildland fires, etc.) are comprised of many contaminates/toxins, therefore it is often difficult (if not impossible) to attribute biological outcomes to any single toxin. In the context of pregnancy exposure, the uteroplacental interface is the absorptive barrier that is relevant to the fetal dose, so the study design (e.g., maternal vs. fetal/offspring focus) and contaminate will largely dictate whether it is possible to assess these “doses.”

Regardless of the study design, studies that do not report a “dose” parameter and only reference the exposure condition, do not allow other investigators (or any subsequent reader) to know the real conditions of the treatment applied, and thus provide limited value for translation and/or comparison to other studies in the field. Also, as previously highlighted (see Pharmacological Properties of Drugs during Pregnancy), it will be important to consider and understand how any physiological changes occurring during pregnancy might influence or affect the dosimetry of environment exposures. For example, pregnancy-related increases in maternal tidal volume (299) will alter normal breathing patterns in a manner that can directly influence the location and amount of PM distributed in the lung (348). Likewise, changes in gastrointestinal mobility, blood acid/base status, and metabolic enzymatic activity are altered during pregnancy (299) and also need to be considered in the context of any relevant environment exposure condition (e.g., inhalation, ingestion, and dermal).

CONCLUSIONS

Recent epidemiological studies highlight the heightened contribution of cardiovascular complications to maternal mortality and morbidity worldwide. This evidence has triggered an increase in research efforts on maternal cardiovascular health during pregnancy and postpartum. In the pursuit of new knowledge and further understanding of maternal cardiac and vascular physiology, it is imperative that the scientific community follows best and standardized practices to conduct research studies in pregnant people and experimental models. The American Journal of Physiology-Heart and Circulatory has previously published guideline articles with step-by-step protocols of how to study cardiac and vascular function in nonpregnant humans and experimental models (for reference, see Table 3) (349355). Pregnancy is a physiological state with unique characteristics of maternal cardiovascular function that dynamically change throughout gestation and postpartum. Hence, our objective was to compliment and extend those previous publications by outlining pregnancy physiology-specific considerations that could lead to appropriate modifications and applications of established experimental protocols. Specifically, we provided theoretical information and practical tips of what factors to consider when studying cardiovascular physiology in pregnant human subjects, experimental animals, or in a dish. Furthermore, we discussed strengths and limitations of various commonly used models of pregnancy-associated diseases and risk factors. Finally, we addressed methodological considerations for the design of rigorous experimental studies in pregnant people and experimental animals. Using these guidelines and underlined best practices, scientists will be empowered to conduct rigorous studies that result in interpretable and translatable outcomes that benefit human maternal health during pregnancy and postpartum.

Table 3.

AJP-Heart and Circulatory references on guidelines for the assessment of cardiac and vascular physiology in nonpregnant experimental models, ex vivo preparations, and cell culture models

Title PMID Citation Year
Guidelines for assessment of cardiac electrophysiology and arrhythmias in small animals 36269644 2022
Guidelines for the measurement of vascular function and structure in isolated arteries and veins 33989082 2021
Guidelines for evaluating myocardial cell death 31418596 2019
Guidelines for authors and reviewers on antibody use in physiology studies 29351459 2018
Guidelines for measuring cardiac physiology in mice 29351456 2018
Guidelines for experimental models of myocardial ischemia and infarction 29351451 2018
Statistical considerations in reporting cardiovascular research 30028200 2018

GRANTS

This work was funded by the following sources: National Institutes of Health Grants R01HL163003 (to H.E.C.), R01HL147844 (to H.E.C.), R01HL163818 (to H.E.C.), R01HL143459 (to B.T.A.), P20GM104357 (to B.T.A.), P20GM121334 (to B.T.A.), R01HL159865 (to M.E.), R01HL131182 (to M.E.), R01HL138181 (to C.G.J.), R01HD088590-06 (to C.G.J.), R21HD111908 (to C.G.J.), P20GM103499 (to H.A.L.), F31ES034646-01 (to I.M.O.), T32ES032920 (to I.M.O.), 1R01HL150472-01A (to E.B.P.), R56HL159447-01 (to J.P.W.), R01HL149608 (to L.Z.), R01HL169157 (to L.Z.), R01HL155295 (to L.Z.), P01HD083132 (to L.Z.), R21NS103017 (to L.Z.), R01HL146562 (to S.G.), and R01HL146562-04S1 (to S.G.); Jewish Heritage fund for excellence faculty support grant (to H.E.C.); National Health and Medical Research Council Ideas Grant APP2002129 (to A.S.C.); National Heart Foundation future leader fellowship (to A.S.C.); Christenson Professor in Active Healthy Living (to M.H.D.); Canadian Institutes of Health Research Foundation grant (to S.T.D.); Distinguished University Professor (to S.T.D.); British Heart Foundation (to D.A.G.) and Grant RG/17/12/33167 (to S.E.O.); Medical Research Council UK (to D.A.G.) and Medical Research Council Grant MC_00014/4 (to S.E.O.); Biotechnology and Biological Sciences Research Council (to D.A.G.); Lister Institute (to D.A.G.), Royal Society (to D.A.G.); Dutch Heart Foundation Dekker Grant 2021T017 (to M.F.H.); U.S. Department of Defense (to C.G.J.); American Heart Association Grant 20CSA35320107 (to I.M.O.), and West Virginia University School of Medicine Synergy Grant Award (to I.M.O.).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

H.E.C., B.T.A., A.S.C., M.H.D., S.T.D., M.E., D.A.G., M.F.H., C.G.J., H.A.L., I.M.O., S.E.O., E.B.P., J.P.W., L.Z., and S.G. conceived and designed research; H.E.C., D.A.G., M.F.H. prepared figures; H.E.C., B.T.A., A.S.C., M.H.D., S.T.D., M.E., D.A.G., M.F.H., C.G.J., H.A.L., I.M.O., S.E.O., E.B.P., J.P.W., L.Z., and S.G. drafted manuscript; H.E.C., B.T.A., A.S.C., M.H.D., S.T.D., M.E., D.A.G., M.F.H., C.G.J., H.A.L., I.M.O., S.E.O., E.B.P., J.P.W., L.Z., and S.G. edited and revised manuscript; H.E.C., B.T.A., A.S.C., M.H.D., S.T.D., M.E., D.A.G., M.F.H., C.G.J., H.A.L., I.M.O., S.E.O., E.B.P., J.P.W., L.Z., and S.G. approved final version of manuscript.

REFERENCES

  • 1. Kuate Defo A, Daskalopoulou SS. Alterations in vessel hemodynamics across uncomplicated pregnancy. Am J Hypertens 36: 183–191, 2023. doi: 10.1093/ajh/hpac132. [DOI] [PubMed] [Google Scholar]
  • 2. Sanghavi M, Rutherford JD. Cardiovascular physiology of pregnancy. Circulation 130: 1003–1008, 2014. doi: 10.1161/CIRCULATIONAHA.114.009029. [DOI] [PubMed] [Google Scholar]
  • 3. Hunter S, Robson SC. Adaptation of the maternal heart in pregnancy. Br Heart J 68: 540–543, 1992. doi: 10.1136/hrt.68.12.540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Sarhaddi F, Azimi I, Axelin A, Niela-Vilen H, Liljeberg P, Rahmani AM. Trends in heart rate and heart rate variability during pregnancy and the 3-month postpartum period: continuous monitoring in a free-living context. JMIR Mhealth Uhealth 10: e33458, 2022. doi: 10.2196/33458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wong AYH, Kulandavelu S, Whiteley KJ, Qu D, Langille BL, Adamson SL. Maternal cardiovascular changes during pregnancy and postpartum in mice. Am J Physiol Heart Circ Physiol 282: H918–H925, 2002. doi: 10.1152/ajpheart.00641.2001. [DOI] [PubMed] [Google Scholar]
  • 6. Clapp JF 3rd, Capeless E. Cardiovascular function before, during, and after the first and subsequent pregnancies. Am J Cardiol 80: 1469–1473, 1997. doi: 10.1016/s0002-9149(97)00738-8. [DOI] [PubMed] [Google Scholar]
  • 7. Tong W, Allison BJ, Brain KL, Patey OV, Niu Y, Botting KJ, Ford SG, Garrud TA, Wooding PFB, Shaw CJ, Lyu Q, Zhang L, Ma J, Cindrova-Davies T, Yung HW, Burton GJ, Giussani DA. Chronic hypoxia in ovine pregnancy recapitulates physiological and molecular markers of preeclampsia in the mother, placenta, and offspring. Hypertension 79: 1525–1535, 2022. doi: 10.1161/HYPERTENSIONAHA.122.19175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Osol G, Mandalà M. Maternal uterine vascular remodeling during pregnancy. Physiology (Bethesda) 24: 58–71, 2009. doi: 10.1152/physiol.00033.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Osol G, Ko NL, Mandalà M. Plasticity of the maternal vasculature during pregnancy. Annu Rev Physiol 81: 89–111, 2019. doi: 10.1146/annurev-physiol-020518-114435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Osol G, Moore LG. Maternal uterine vascular remodeling during pregnancy. Microcirculation 21: 38–47, 2014. doi: 10.1111/micc.12080. [DOI] [PubMed] [Google Scholar]
  • 11. Chung E, Leinwand LA. Pregnancy as a cardiac stress model. Cardiovasc Res 101: 561–570, 2014. doi: 10.1093/cvr/cvu013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Eghbali M, Deva R, Alioua A, Minosyan TY, Ruan H, Wang Y, Toro L, Stefani E. Molecular and functional signature of heart hypertrophy during pregnancy. Circ Res 96: 1208–1216, 2005. doi: 10.1161/01.RES.0000170652.71414.16. [DOI] [PubMed] [Google Scholar]
  • 13. Fulghum KL, Smith JB, Chariker J, Garrett LF, Brittian KR, Lorkiewicz PK, McNally LA, Uchida S, Jones SP, Hill BG, Collins HE. Metabolic signatures of pregnancy-induced cardiac growth. Am J Physiol Heart Circ Physiol 323: H146–H164, 2022. doi: 10.1152/ajpheart.00105.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Liu LX, Rowe GC, Yang S, Li J, Damilano F, Chan MC, Lu W, Jang C, Wada S, Morley M, Hesse M, Fleischmann BK, Rabinowitz JD, Das S, Rosenzweig A, Arany Z. PDK4 inhibits cardiac pyruvate oxidation in late pregnancy. Circ Res 121: 1370–1378, 2017. doi: 10.1161/CIRCRESAHA.117.311456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Schulman-Geltzer EB, Fulghum KL, Singhal RA, Hill BG, Collins HE. Cardiac mitochondrial metabolism during pregnancy and the postpartum period. Am J Physiol Heart Circ Physiol 326: H1324–H1335, 2024. doi: 10.1152/ajpheart.00127.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Jellyman JK, Gardner DS, Edwards CM, Fowden AL, Giussani DA. The effects of pregnancy on the cardiovascular response to acute systemic isocapnic hypoxia in conscious sheep. BJOG 112: 889–896, 2005. doi: 10.1111/j.1471-0528.2005.00570.x. [DOI] [PubMed] [Google Scholar]
  • 17. Chung E, Heimiller J, Leinwand LA. Distinct cardiac transcriptional profiles defining pregnancy and exercise. PLoS One 7: e42297, 2012. doi: 10.1371/journal.pone.0042297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Chung E, Yeung F, Leinwand LA. Calcineurin activity is required for cardiac remodelling in pregnancy. Cardiovasc Res 100: 402–410, 2013. doi: 10.1093/cvr/cvt208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Chung E, Yeung F, Leinwand LA. Akt and MAPK signaling mediate pregnancy-induced cardiac adaptation. J Appl Physiol (1985) 112: 1564–1575, 2012. doi: 10.1152/japplphysiol.00027.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Ampey BC, Ampey AC, Lopez GE, Bird IM, Magness RR. Cyclic nucleotides differentially regulate Cx43 gap junction function in uterine artery endothelial cells from pregnant ewes. Hypertension 70: 401–411, 2017. doi: 10.1161/HYPERTENSIONAHA.117.09113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Li Y, Zheng J, Bird IM, Magness RR. Effects of pulsatile shear stress on nitric oxide production and endothelial cell nitric oxide synthase expression by ovine fetoplacental artery endothelial cells. Biol Reprod 69: 1053–1059, 2003. doi: 10.1095/biolreprod.102.013474. [DOI] [PubMed] [Google Scholar]
  • 22. Rupnow HL, Phernetton TM, Modrick ML, Wiltbank MC, Bird IM, Magness RR. Endothelial vasodilator production by uterine and systemic arteries. VIII. Estrogen and progesterone effects on cPLA2, COX-1, and PGIS protein expression. Biol Reprod 66: 468–474, 2002. doi: 10.1095/biolreprod66.2.468. [DOI] [PubMed] [Google Scholar]
  • 23. Song R, Hu X-Q, Romero M, Holguin MA, Kagabo W, Xiao D, Wilson SM, Zhang L. Ryanodine receptor subtypes regulate Ca2+ sparks/spontaneous transient outward currents and myogenic tone of uterine arteries in pregnancy. Cardiovasc Res 117: 792–804, 2021. doi: 10.1093/cvr/cvaa089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Mukhtarova N, Ko NL, Gokina NI, Mandalà M, Osol G. Enhanced vascular smooth muscle calcium sensitivity and loss of endothelial vasodilator influence contribute to myogenic tone development in rat radial uterine arteries during gestation. J Vasc Res 57: 126–135, 2020. doi: 10.1159/000505670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Hu X-Q, Song R, Romero M, Dasgupta C, Huang X, Holguin MA, Williams V, Xiao D, Wilson SM, Zhang L. Pregnancy increases Ca(2+) sparks/spontaneous transient outward currents and reduces uterine arterial myogenic tone. Hypertension 73: 691–702, 2019. doi: 10.1161/HYPERTENSIONAHA.118.12484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Eckman DM, Gupta R, Rosenfeld CR, Morgan TM, Charles SM, Mertz H, Moore LG. Pregnancy increases myometrial artery myogenic tone via NOS- or COX-independent mechanisms. Am J Physiol Regul Integr Comp Physiol 303: R368–R375, 2012. doi: 10.1152/ajpregu.00490.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Veerareddy S, Cooke CL, Baker PN, Davidge ST. Vascular adaptations to pregnancy in mice: effects on myogenic tone. Am J Physiol Heart Circ Physiol 283: H2226–H2233, 2002. doi: 10.1152/ajpheart.00593.2002. [DOI] [PubMed] [Google Scholar]
  • 28. Mann SE, Maille N, Clas D, Osol G. Perivascular adipose tissue: a novel regulator of vascular tone in the rat pregnancy. Reprod Sci 22: 802–807, 2015. doi: 10.1177/1933719114561556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Osikoya O, Ahmed H, Panahi S, Bourque SL, Goulopoulou S. Uterine perivascular adipose tissue is a novel mediator of uterine artery blood flow and reactivity in rat pregnancy. J Physiol 597: 3833–3852, 2019. doi: 10.1113/JP277643. [DOI] [PubMed] [Google Scholar]
  • 30. Osikoya O, Cushen SC, Gardner JJ, Raetz MM, Nagarajan B, Raut S, Goulopoulou S. Exosomes facilitate intercellular communication between uterine perivascular adipose tissue and vascular smooth muscle cells in pregnant rats. Am J Physiol Heart Circ Physiol 323: H577–H584, 2022. doi: 10.1152/ajpheart.00322.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Bernstein IM, Ziegler WF, Leavitt T, Badger GJ. Uterine artery hemodynamic adaptations through the menstrual cycle into early pregnancy. Obstet Gynecol 99: 620–624, 2002. doi: 10.1016/s0029-7844(01)01787-2. [DOI] [PubMed] [Google Scholar]
  • 32. Palmer SK, Zamudio S, Coffin C, Parker S, Stamm E, Moore LG. Quantitative estimation of human uterine artery blood flow and pelvic blood flow redistribution in pregnancy. Obstet Gynecol 80: 1000–1006, 1992. [PubMed] [Google Scholar]
  • 33. Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 99: 196–211, 2018. doi: 10.1093/biolre/ioy049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Ko NL, Mandalà M, John L, Gelinne A, Osol G. Venoarterial communication mediates arterial wall shear stress-induced maternal uterine vascular remodeling during pregnancy. Am J Physiol Heart Circ Physiol 315: H709–H717, 2018. doi: 10.1152/ajpheart.00126.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. John L, Ko NL, Gokin A, Gokina N, Mandalà M, Osol G. The Piezo1 cation channel mediates uterine artery shear stress mechanotransduction and vasodilation during rat pregnancy. Am J Physiol Heart Circ Physiol 315: H1019–H1026, 2018. doi: 10.1152/ajpheart.00103.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Bulmer JN, Innes BA, Levey J, Robson SC, Lash GE. The role of vascular smooth muscle cell apoptosis and migration during uterine spiral artery remodeling in normal human pregnancy. FASEB J 26: 2975–2985, 2012. doi: 10.1096/fj.12-203679. [DOI] [PubMed] [Google Scholar]
  • 37. Allerkamp HH, Leighton SC, Pole T, Clark AR, James JL. Synergistic regulation of uterine radial artery adaptation to pregnancy by paracrine and haemodynamic factors. Am J Physiol Heart Circ Physiol 325: H790–H805, 2023. doi: 10.1152/ajpheart.00205.2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Allerkamp HH, Pole T, Boukham A, James JL, Clark AR. Pregnancy-specific uterine vascular reactivity: a data-driven computational model of shear-dependent, myogenic, and mechanical radial artery features. Am J Physiol Heart Circ Physiol 323: H72–H88, 2022. doi: 10.1152/ajpheart.00693.2021. [DOI] [PubMed] [Google Scholar]
  • 39. Lorca RA, Wakle-Prabagaran M, Freeman WE, Pillai MK, England SK. The large-conductance voltage- and Ca(2+)-activated K(+) channel and its gamma1-subunit modulate mouse uterine artery function during pregnancy. J Physiol 596: 1019–1033, 2018. doi: 10.1113/JP274524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Spaans F, Quon A, Rowe SR, Morton JS, Kirschenman R, Sawamura T, Tannetta DS, Sargent IL, Davidge ST. Alterations in vascular function by syncytiotrophoblast extracellular vesicles via lectin-like oxidized low-density lipoprotein receptor-1 in mouse uterine arteries. Clin Sci (Lond) 132: 2369–2381, 2018. doi: 10.1042/CS20180639. [DOI] [PubMed] [Google Scholar]
  • 41. Dickey RP, Hower JF. Ultrasonographic features of uterine blood flow during the first 16 weeks of pregnancy. Hum Reprod 10: 2448–2452, 1995. doi: 10.1093/oxfordjournals.humrep.a136317. [DOI] [PubMed] [Google Scholar]
  • 42. Collins SL, Grant D, Black RS, Vellayan M, Impey L. Abdominal pregnancy: a perfusion confusion? Placenta 32: 793–795, 2011. doi: 10.1016/j.placenta.2011.07.032. [DOI] [PubMed] [Google Scholar]
  • 43. Furukawa S, Kuroda Y, Sugiyama A. A comparison of the histological structure of the placenta in experimental animals. J Toxicol Pathol 27: 11–18, 2014. [Erratum in J Toxicol Pathol 29: 74, 2016]. doi: 10.1293/tox.2013-0060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Leiser R, Kaufmann P. Placental structure: in a comparative aspect. Exp Clin Endocrinol 102: 122–134, 1994. doi: 10.1055/s-0029-1211275. [DOI] [PubMed] [Google Scholar]
  • 45. Torgersen KL, Curran CA. A systematic approach to the physiologic adaptations of pregnancy. Crit Care Nurs Q 29: 2–19, 2006. doi: 10.1097/00002727-200601000-00002. [DOI] [PubMed] [Google Scholar]
  • 46. Johnson AC, Cipolla MJ. The cerebral circulation during pregnancy: adapting to preserve normalcy. Physiology (Bethesda) 30: 139–147, 2015. doi: 10.1152/physiol.00048.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Murtada SI, Latorre M, Humphrey JD. Remodeling of the uterine artery during and early after pregnancy in the mouse. Biomech Model Mechanobiol 22: 1531–1540, 2022. doi: 10.1007/s10237-022-01674-2. [DOI] [PubMed] [Google Scholar]
  • 48. Mosca L, Benjamin EJ, Berra K, Bezanson JL, Dolor RJ, Lloyd-Jones DM, , et al. Effectiveness-based guidelines for the prevention of cardiovascular disease in women—2011 update: a guideline from the american heart association. Circulation 123: 1243–1262, 2011. [Erratum in Circulation 123: e624, 2011; Circulation 124: e427, 2011]. doi: 10.1161/CIR.0b013e31820faaf8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Tan EK, Tan EL. Alterations in physiology and anatomy during pregnancy. Best Pract Res Clin Obstet Gynaecol 27: 791–802, 2013. doi: 10.1016/j.bpobgyn.2013.08.001. [DOI] [PubMed] [Google Scholar]
  • 50. Dimitriadis E, Rolnik DL, Zhou W, Estrada-Gutierrez G, Koga K, Francisco RPV, Whitehead C, Hyett J, da Silva Costa F, Nicolaides K, Menkhorst E. Pre-eclampsia. Nat Rev Dis Primers 9: 8, 2023. [Erratum in Nat Rev Dis Primers 9: 35, 2023]. doi: 10.1038/s41572-023-00417-6. [DOI] [PubMed] [Google Scholar]
  • 51. Badrov MB, Park SY, Yoo J-K, Hieda M, Okada Y, Jarvis SS, Stickford AS, Best SA, Nelson DB, Fu Q. Role of corin in blood pressure regulation in normotensive and hypertensive pregnancy. Hypertension 73: 432–439, 2019. doi: 10.1161/HYPERTENSIONAHA.118.12137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Thijssen DHJ, Bruno RM, van Mil ACCM, Holder SM, Faita F, Greyling A, Zock PL, Taddei S, Deanfield JE, Luscher T, Green DJ, Ghiadoni L. Expert consensus and evidence-based recommendations for the assessment of flow-mediated dilation in humans. Eur Heart J 40: 2534–2547, 2019. doi: 10.1093/eurheartj/ehz350. [DOI] [PubMed] [Google Scholar]
  • 53. Countouris ME, Schwarz EB, Rossiter BC, Althouse AD, Berlacher KL, Jeyabalan A, Catov JM. Effects of lactation on postpartum blood pressure among women with gestational hypertension and preeclampsia. Am J Obstet Gynecol 215: 241 e241–241.e8, 2016. doi: 10.1016/j.ajog.2016.02.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Faulkner JL, Wright D, Antonova G, Jaffe IZ, Kennard S, Belin de Chantemèle EJ. Midgestation leptin infusion induces characteristics of clinical preeclampsia in mice, which is ablated by endothelial mineralocorticoid receptor deletion. Hypertension 79: 1536–1547, 2022. doi: 10.1161/HYPERTENSIONAHA.121.18832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Beeson JH, Blackmore HL, Carr SK, Dearden L, Duque-Guimarães DE, Kusinski LC, Pantaleão LC, Pinnock AG, Aiken CE, Giussani DA, Fernandez-Twinn DS, Ozanne SE. Maternal exercise intervention in obese pregnancy improves the cardiovascular health of the adult male offspring. Mol Metab 16: 35–44, 2018. doi: 10.1016/j.molmet.2018.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Zhao X, Sun Y, Zhang H, Zhang Y, Zhao H, Yao X, Zhang W. Effect of different iodide intake during pregnancy and lactation on thyroid and cardiovascular function in maternal and offspring rats. J Trace Elem Med Biol 79: 127267, 2023. doi: 10.1016/j.jtemb.2023.127267. [DOI] [PubMed] [Google Scholar]
  • 57. Abdelnaby EA, Yasin NAE, Abouelela YS, Rashad E, Daghash SM, El-Sherbiny HR. Ovarian, uterine, and luteal vascular perfusions during follicular and luteal phases in the adult cyclic female rabbits with special orientation to their histological detection of hormone receptor. BMC Vet Res 18: 301, 2022. doi: 10.1186/s12917-022-03390-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Morrison JL, Botting KJ, Darby JRT, David AL, Dyson RM, Gatford KL, Gray C, Herrera EA, Hirst JJ, Kim B, Kind KL, Krause BJ, Matthews SG, Palliser HK, Regnault TRH, Richardson BS, Sasaki A, Thompson LP, Berry MJ. Guinea pig models for translation of the developmental origins of health and disease hypothesis into the clinic. J Physiol 596: 5535–5569, 2018. doi: 10.1113/JP274948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Kuo AH, Li C, Li J, Huber HF, Nathanielsz PW, Clarke GD. Cardiac remodelling in a baboon model of intrauterine growth restriction mimics accelerated ageing. J Physiol 595: 1093–1110, 2017. doi: 10.1113/JP272908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Barker SJ, Gamel DM, Tremper KK. Cardiovascular effects of anesthesia and operation. Crit Care Clin 3: 251–268, 1987. [PubMed] [Google Scholar]
  • 61. Khan I, Dekou V, Hanson M, Poston L, Taylor P. Predictive adaptive responses to maternal high-fat diet prevent endothelial dysfunction but not hypertension in adult rat offspring. Circulation 110: 1097–1102, 2004. doi: 10.1161/01.CIR.0000139843.05436.A0. [DOI] [PubMed] [Google Scholar]
  • 62. Spiroski A-M, Niu Y, Nicholas LM, Austin-Williams S, Camm EJ, Sutherland MR, Ashmore TJ, Skeffington KL, Logan A, Ozanne SE, Murphy MP, Giussani DA. Mitochondria antioxidant protection against cardiovascular dysfunction programmed by early-onset gestational hypoxia. FASEB J 35: e21446, 2021. doi: 10.1096/fj.202002705R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Kane AD, Herrera EA, Camm EJ, Giussani DA. Vitamin C prevents intrauterine programming of in vivo cardiovascular dysfunction in the rat. Circ J 77: 2604–2611, 2013. doi: 10.1253/circj.cj-13-0311. [DOI] [PubMed] [Google Scholar]
  • 64. Lakshman R, Spiroski AM, McIver LB, Murphy MP, Giussani DA. Noninvasive biomarkers for cardiovascular dysfunction programmed in male offspring of adverse pregnancy. Hypertension 78: 1818–1828, 2021. doi: 10.1161/HYPERTENSIONAHA.121.17926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Nuzzo AM, Camm EJ, Sferruzzi-Perri AN, Ashmore TJ, Yung H-W, Cindrova-Davies T, Spiroski A-M, Sutherland MR, Logan A, Austin-Williams S, Burton GJ, Rolfo A, Todros T, Murphy MP, Giussani DA. Placental adaptation to early-onset hypoxic pregnancy and mitochondria-targeted antioxidant therapy in a rodent model. Am J Pathol 188: 2704–2716, 2018. doi: 10.1016/j.ajpath.2018.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Richter HG, Camm EJ, Modi BN, Naeem F, Cross CM, Cindrova-Davies T, Spasic-Boskovic O, Dunster C, Mudway IS, Kelly FJ, Burton GJ, Poston L, Giussani DA. Ascorbate prevents placental oxidative stress and enhances birth weight in hypoxic pregnancy in rats. J Physiol 590: 1377–1387, 2012. doi: 10.1113/jphysiol.2011.226340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Zhang Y, Wu J, King JH, Huang CL, Fraser JA. Measurement and interpretation of electrocardiographic QT intervals in murine hearts. Am J Physiol Heart Circ Physiol 306: H1553–H1557, 2014. doi: 10.1152/ajpheart.00459.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Blackmore HL, Niu Y, Fernandez-Twinn DS, Tarry-Adkins JL, Giussani DA, Ozanne SE. Maternal diet-induced obesity programs cardiovascular dysfunction in adult male mouse offspring independent of current body weight. Endocrinology 155: 3970–3980, 2014. doi: 10.1210/en.2014-1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Skeffington KL, Higgins JS, Mahmoud AD, Evans AM, Sferruzzi-Perri AN, Fowden AL, Yung HW, Burton GJ, Giussani DA, Moore LG. Hypoxia, AMPK activation and uterine artery vasoreactivity. J Physiol 594: 1357–1369, 2016. doi: 10.1113/JP270995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Villalobos-Labra R, Liu R, Spaans F, Sáez T, Semeria Maitret T, Quon A, Sawamura T, Cooke C-LM, Davidge ST. Placenta-derived extracellular vesicles from preeclamptic pregnancies impair vascular endothelial function via lectin-like oxidized LDL receptor-1. Hypertension 80: 2226–2238, 2023. doi: 10.1161/HYPERTENSIONAHA.123.21205. [DOI] [PubMed] [Google Scholar]
  • 71. Soares MJ, Chakraborty D, Karim Rumi MA, Konno T, Renaud SJ. Rat placentation: an experimental model for investigating the hemochorial maternal-fetal interface. Placenta 33: 233–243, 2012. doi: 10.1016/j.placenta.2011.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Rueda-Clausen CF, Morton JS, Davidge ST. The early origins of cardiovascular health and disease: who, when, and how. Semin Reprod Med 29: 197–210, 2011. doi: 10.1055/s-0031-1275520. [DOI] [PubMed] [Google Scholar]
  • 73. Shaw CJ, Rivens I, Civale J, Botting KJ, Allison BJ, Brain KL, Niu Y, Ter Haar G, Giussani DA, Lees CC. Maternal and fetal cardiometabolic recovery following ultrasound-guided high-intensity focused ultrasound placental vascular occlusion. J R Soc Interface 16: 20190013, 2019. doi: 10.1098/rsif.2019.0013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Allison BJ, Brain KL, Niu Y, Kane AD, Herrera EA, Thakor AS, Botting KJ, Cross CM, Itani N, Shaw CJ, Skeffington KL, Beck C, Giussani DA. Altered cardiovascular defense to hypotensive stress in the chronically hypoxic fetus. Hypertension 76: 1195–1207, 2020. doi: 10.1161/HYPERTENSIONAHA.120.15384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Allison BJ, Brain KL, Niu Y, Kane AD, Herrera EA, Thakor AS, Botting KJ, Cross CM, Itani N, Skeffington KL, Beck C, Giussani DA. Fetal in vivo continuous cardiovascular function during chronic hypoxia. J Physiol 594: 1247–1264, 2016. doi: 10.1113/JP271091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Giussani DA. Breath of life: heart disease link to developmental hypoxia. Circulation 144: 1429–1443, 2021. doi: 10.1161/CIRCULATIONAHA.121.054689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Wooding P, Burton G. Comparative Placentation: Structures, Functions and Evolution (1st ed.). Heidelberg: Springer Berlin, 2008. [Google Scholar]
  • 78. Schröder M, Kaufman RJ. The mammalian unfolded protein response. Annu Rev Biochem 74: 739–789, 2005. doi: 10.1146/annurev.biochem.73.011303.074134. [DOI] [PubMed] [Google Scholar]
  • 79. Hu X-Q, Dasgupta C, Xiao D, Huang X, Yang S, Zhang L. MicroRNA-210 targets ten-eleven translocation methylcytosine dioxygenase 1 and suppresses pregnancy-mediated adaptation of large conductance Ca(2+)-activated K(+) channel expression and function in ovine uterine arteries. Hypertension, 2017. doi: 10.1161/HYPERTENSIONAHA.117.09864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Llanos AJ, Riquelme RA, Sanhueza EM, Hanson MA, Blanco CE, Parer JT, Herrera EA, Pulgar VM, Reyes RV, Cabello G, Giussani DA. The fetal llama versus the fetal sheep: different strategies to withstand hypoxia. High Alt Med Biol 4: 193–202, 2003. doi: 10.1089/152702903322022794. [DOI] [PubMed] [Google Scholar]
  • 81. Frias AE, Morgan TK, Evans AE, Rasanen J, Oh KY, Thornburg KL, Grove KL. Maternal high-fat diet disturbs uteroplacental hemodynamics and increases the frequency of stillbirth in a nonhuman primate model of excess nutrition. Endocrinology 152: 2456–2464, 2011. doi: 10.1210/en.2010-1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Denney JM, Bird C, Gendron-Fitzpatrick A, Sampene E, Bird IM, Shah DM. Renin-angiotensin system transgenic mouse model recapitulates pathophysiology similar to human preeclampsia with renal injury that may be mediated through VEGF. Am J Physiol Renal Physiol 312: F445–F455, 2017. doi: 10.1152/ajprenal.00108.2016. [DOI] [PubMed] [Google Scholar]
  • 83. Mirabito KM, Hilliard LM, Wei Z, Tikellis C, Widdop RE, Vinh A, Denton KM. Role of inflammation and the angiotensin type 2 receptor in the regulation of arterial pressure during pregnancy in mice. Hypertension 64: 626–631, 2014. doi: 10.1161/HYPERTENSIONAHA.114.03189. [DOI] [PubMed] [Google Scholar]
  • 84. Burke SD, Barrette VF, David S, Khankin EV, Adams MA, Croy BA. Circulatory and renal consequences of pregnancy in diabetic NOD mice. Placenta 32: 949–955, 2011. doi: 10.1016/j.placenta.2011.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Burke SD, Barrette VF, Bianco J, Thorne JG, Yamada AT, Pang SC, Adams MA, Croy BA. Spiral arterial remodeling is not essential for normal blood pressure regulation in pregnant mice. Hypertension 55: 729–737, 2010. doi: 10.1161/HYPERTENSIONAHA.109.144253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Butz GM, Davisson RL. Long-term telemetric measurement of cardiovascular parameters in awake mice: a physiological genomics tool. Physiol Genomics 5: 89–97, 2001. doi: 10.1152/physiolgenomics.2001.5.2.89. [DOI] [PubMed] [Google Scholar]
  • 87. Kane AD, Niu Y, Herrera EA, Morton AJ, Giussani DA. Impaired nitric oxide mediated vasodilation in the peripheral circulation in the R6/2 mouse model of Huntington’s disease. Sci Rep 6: 25979, 2016. doi: 10.1038/srep25979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Niu Y, Kane AD, Lusby CM, Allison BJ, Chua YY, Kaandorp JJ, Nevin-Dolan R, Ashmore TJ, Blackmore HL, Derks JB, Ozanne SE, Giussani DA. Maternal allopurinol prevents cardiac dysfunction in adult male offspring programmed by chronic hypoxia during pregnancy. Hypertension 72: 971–978, 2018. doi: 10.1161/HYPERTENSIONAHA.118.11363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Hansell JA, Richter HG, Camm EJ, Herrera EA, Blanco CE, Villamor E, Patey OV, Lock MC, Trafford AW, Galli GLJ, Giussani DA. Maternal melatonin: effective intervention against developmental programming of cardiovascular dysfunction in adult offspring of complicated pregnancy. J Pineal Res 72: e12766, 2022. doi: 10.1111/jpi.12766. [DOI] [PubMed] [Google Scholar]
  • 90. Vaughan OR, Rossi CA, Ginsberg Y, White A, Hristova M, Sebire NJ, Martin J, Zachary IC, Peebles DM, David AL. Perinatal and long-term effects of maternal uterine artery adenoviral VEGF-A165 gene therapy in the growth-restricted guinea pig fetus. Am J Physiol Regul Integr Comp Physiol 315: R344–R353, 2018. doi: 10.1152/ajpregu.00210.2017. [DOI] [PubMed] [Google Scholar]
  • 91. Haq KT, Cooper BL, Berk F, Posnack NG. The effect of sex and age on ex vivo cardiac electrophysiology: insight from a guinea pig model. Am J Physiol Heart Circ Physiol 324: H141–H154, 2023. doi: 10.1152/ajpheart.00497.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Herrera EA, Cifuentes-Zúñiga F, Figueroa E, Villanueva C, Hernández C, Alegría R, Arroyo-Jousse V, Peñaloza E, Farías M, Uauy R, Casanello P, Krause BJ. N-Acetylcysteine, a glutathione precursor, reverts vascular dysfunction and endothelial epigenetic programming in intrauterine growth restricted guinea pigs. J Physiol 595: 1077–1092, 2017. doi: 10.1113/JP273396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Hargrave B, Lattanzio F. Cocaine activates the renin-angiotensin system in pregnant rabbits and alters the response to ischemia. Cardiovasc Toxicol 2: 91–97, 2002. doi: 10.1385/ct:2:2:091. [DOI] [PubMed] [Google Scholar]
  • 94. Khammy MM, Angus JA, Wright CE. Vascular reactivity of rabbit isolated renal and femoral resistance arteries in renal wrap hypertension. Eur J Pharmacol 773: 32–41, 2016. doi: 10.1016/j.ejphar.2016.01.004. [DOI] [PubMed] [Google Scholar]
  • 95. Anwar MA, Ju K, Docherty CC, Poston L, Nathanielsz PW. Developmental changes in reactivity of small femoral arteries in the fetal and postnatal baboon. Am J Obstet Gynecol 184: 707–712, 2001. doi: 10.1067/mob.2001.111159. [DOI] [PubMed] [Google Scholar]
  • 96. Herrera EA, Reyes RV, Giussani DA, Riquelme RA, Sanhueza EM, Ebensperger G, Casanello P, Méndez N, Ebensperger R, Sepúlveda-Kattan E, Pulgar VM, Cabello G, Blanco CE, Hanson MA, Parer JT, Llanos AJ. Carbon monoxide: a novel pulmonary artery vasodilator in neonatal llamas of the Andean altiplano. Cardiovasc Res 77: 197–201, 2008. doi: 10.1093/cvr/cvm013. [DOI] [PubMed] [Google Scholar]
  • 97. Nathanielsz PW, Jenkins SL, Tame JD, Winter JA, Guller S, Giussani DA. Local paracrine effects of estradiol are central to parturition in the rhesus monkey. Nat Med 4: 456–459, 1998. doi: 10.1038/nm0498-456. [DOI] [PubMed] [Google Scholar]
  • 98. Giussani DA, Jenkins SL, Mecenas CA, Winter JA, Honnebier BO, Wu W, Nathanielsz PW. Daily and hourly temporal association between delta4-androstenedione-induced preterm myometrial contractions and maternal plasma estradiol and oxytocin concentrations in the 0.8 gestation rhesus monkey. Am J Obstet Gynecol 174: 1050–1055, 1996. doi: 10.1016/s0002-9378(96)70349-6. [DOI] [PubMed] [Google Scholar]
  • 99. Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, Lan F, Diecke S, Huber B, Mordwinkin NM, Plews JR, Abilez OJ, Cui B, Gold JD, Wu JC. Chemically defined generation of human cardiomyocytes. Nat Methods 11: 855–860, 2014. doi: 10.1038/nmeth.2999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Dunn KK, Reichardt IM, Simmons AD, Jin G, Floy ME, Hoon KM, Palecek SP. Coculture of endothelial cells with human pluripotent stem cell-derived cardiac progenitors reveals a differentiation stage-specific enhancement of cardiomyocyte maturation. Biotechnol J 14: e1800725, 2019. doi: 10.1002/biot.201800725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Beauchamp P, Jackson CB, Ozhathil LC, Agarkova I, Galindo CL, Sawyer DB, Suter TM, Zuppinger C. 3D co-culture of hiPSC-derived cardiomyocytes with cardiac fibroblasts improves tissue-like features of cardiac spheroids. Front Mol Biosci 7: 14, 2020. doi: 10.3389/fmolb.2020.00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Zimmermann W-H, Schneiderbanger K, Schubert P, Didié M, Münzel F, Heubach JF, Kostin S, Neuhuber WL, Eschenhagen T. Tissue engineering of a differentiated cardiac muscle construct. Circ Res 90: 223–230, 2002. doi: 10.1161/hh0202.103644. [DOI] [PubMed] [Google Scholar]
  • 103. Ronaldson-Bouchard K, Ma SP, Yeager K, Chen T, Song LJin, Sirabella D, Morikawa K, Teles D, Yazawa M, Vunjak-Novakovic G. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556: 239–243, 2018. [Erratum in Nature 572: E16–E17, 2019]. doi: 10.1038/s41586-018-0016-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Schock H, Zeleniuch-Jacquotte A, Lundin E, Grankvist K, Lakso H-Å, Idahl A, Lehtinen M, Surcel H-M, Fortner RT. Hormone concentrations throughout uncomplicated pregnancies: a longitudinal study. BMC Pregnancy Childbirth 16: 146, 2016. doi: 10.1186/s12884-016-0937-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Chen L, Mir SA, Bendt AK, Chua EWL, Narasimhan K, Tan KM-L, Loy SL, Tan KH, Shek LP, Chan J, Yap F, Meaney MJ, Chan S-Y, Chong YS, Gluckman PD, Eriksson JG, Karnani N, Wenk MR. Plasma lipidomic profiling reveals metabolic adaptations to pregnancy and signatures of cardiometabolic risk: a preconception and longitudinal cohort study. BMC Med 21: 53, 2023. doi: 10.1186/s12916-023-02740-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Di Cianni G, Miccoli R, Volpe L, Lencioni C, Del Prato S. Intermediate metabolism in normal pregnancy and in gestational diabetes. Diabetes Metab Res Rev 19: 259–270, 2003. doi: 10.1002/dmrr.390. [DOI] [PubMed] [Google Scholar]
  • 107. Noshiro K, Umazume T, Hattori R, Kataoka S, Yamada T, Watari H. Changes in serum levels of ketone bodies and human chorionic gonadotropin during pregnancy in relation to the neonatal body shape: a retrospective analysis. Nutrients 14: 1971, 2022. doi: 10.3390/nu14091971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. de Haas S, Mulder E, Schartmann N, Mohseni Z, Abo Hasson F, Alsadah F, van Kuijk S, van Drongelen J, Ghossein-Doha C, Spaanderman M. Blood pressure adjustments throughout healthy and hypertensive pregnancy: a systematic review and meta-analysis. Pregnancy Hypertens 27: 51–58, 2022. doi: 10.1016/j.preghy.2021.12.004. [DOI] [PubMed] [Google Scholar]
  • 109. de Haas S, Ghossein-Doha C, van Kuijk SM, van Drongelen J, Spaanderman ME. Physiological adaptation of maternal plasma volume during pregnancy: a systematic review and meta-analysis. Ultrasound Obstet Gynecol 49: 177–187, 2017. doi: 10.1002/uog.17360. [DOI] [PubMed] [Google Scholar]
  • 110. Pillekamp F, Haustein M, Khalil M, Emmelheinz M, Nazzal R, Adelmann R, Nguemo F, Rubenchyk O, Pfannkuche K, Matzkies M, Reppel M, Bloch W, Brockmeier K, Hescheler J. Contractile properties of early human embryonic stem cell-derived cardiomyocytes: beta-adrenergic stimulation induces positive chronotropy and lusitropy but not inotropy. Stem Cells Dev 21: 2111–2121, 2012. doi: 10.1089/scd.2011.0312. [DOI] [PubMed] [Google Scholar]
  • 111. Liu J, Sun N, Bruce MA, Wu JC, Butte MJ. Atomic force mechanobiology of pluripotent stem cell-derived cardiomyocytes. PLoS One 7: e37559, 2012. doi: 10.1371/journal.pone.0037559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Hirt MN, Boeddinghaus J, Mitchell A, Schaaf S, Börnchen C, Müller C, Schulz H, Hubner N, Stenzig J, Stoehr A, Neuber C, Eder A, Luther PK, Hansen A, Eschenhagen T. Functional improvement and maturation of rat and human engineered heart tissue by chronic electrical stimulation. J Mol Cell Cardiol 74: 151–161, 2014. doi: 10.1016/j.yjmcc.2014.05.009. [DOI] [PubMed] [Google Scholar]
  • 113. Martherus RSRM, Vanherle SJV, Timmer EDJ, Zeijlemaker VA, Broers JL, Smeets HJ, Geraedts JP, Ayoubi TAY. Electrical signals affect the cardiomyocyte transcriptome independently of contraction. Physiol Genomics 42A: 283–289, 2010. doi: 10.1152/physiolgenomics.00182.2009. [DOI] [PubMed] [Google Scholar]
  • 114. Godier-Furnémont AFG, Tiburcy M, Wagner E, Dewenter M, Lämmle S, El-Armouche A, Lehnart SE, Vunjak-Novakovic G, Zimmermann W-H. Physiologic force-frequency response in engineered heart muscle by electromechanical stimulation. Biomaterials 60: 82–91, 2015. doi: 10.1016/j.biomaterials.2015.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Ovchinnikova E, Hoes M, Ustyantsev K, Bomer N, de Jong TV, van der Mei H, Berezikov E, van der Meer P. Modeling human cardiac hypertrophy in stem cell-derived cardiomyocytes. Stem Cell Reports 10: 794–807, 2018. doi: 10.1016/j.stemcr.2018.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Rysä J, Tokola H, Ruskoaho H. Mechanical stretch induced transcriptomic profiles in cardiac myocytes. Sci Rep 8: 4733, 2018. doi: 10.1038/s41598-018-23042-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Bliley JM, Vermeer MCSC, Duffy RM, Batalov I, Kramer D, Tashman JW, Shiwarski DJ, Lee A, Teplenin AS, Volkers L, Coffin B, Hoes MF, Kalmykov A, Palchesko RN, Sun Y, Jongbloed JDH, Bomer N, de Boer RA, Suurmeijer AJH, Pijnappels DA, Bolling MC, van der Meer P, Feinberg AW. Dynamic loading of human engineered heart tissue enhances contractile function and drives a desmosome-linked disease phenotype. Sci Transl Med 13: eabd1817, 2021. doi: 10.1126/scitranslmed.abd1817. [DOI] [PubMed] [Google Scholar]
  • 118.GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 396: 1204–1222, 2020. [Erratum in Lancet 396: 1562, 2020]. doi: 10.1016/S0140-6736(20)30925-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Gestational Hypertension and Preeclampsia: ACOG Practice Bulletin Summary, Number 222. Obstet Gynecol 135: 1492–1495, 2020. doi: 10.1097/AOG.0000000000003892. [DOI] [PubMed] [Google Scholar]
  • 120. Brown MA, Magee LA, Kenny LC, Karumanchi SA, McCarthy FP, Saito S, Hall DR, Warren CE, Adoyi G, Ishaku S; International Society for the Study of Hypertension in Pregnancy (ISSHP). Hypertensive disorders of pregnancy: ISSHP classification, diagnosis, and management recommendations for international practice. Hypertension 72: 24–43, 2018. doi: 10.1161/HYPERTENSIONAHA.117.10803. [DOI] [PubMed] [Google Scholar]
  • 121.World Health Organization. WHO Recommendations for Prevention and Treatment of Pre-eclampsia and Eclampsia. Geneva: World Health Organization, 2011. [PubMed] [Google Scholar]
  • 122. Cushen SC, Goulopoulou S. New models of pregnancy-associated hypertension. Am J Hypertens 30: 1053–1062, 2017. doi: 10.1093/ajh/hpx063. [DOI] [PubMed] [Google Scholar]
  • 123. Intapad S, Warrington JP, Spradley FT, Palei AC, Drummond HA, Ryan MJ, Granger JP, Alexander BT. Reduced uterine perfusion pressure induces hypertension in the pregnant mouse. Am J Physiol Regul Integr Comp Physiol 307: R1353–R1357, 2014. doi: 10.1152/ajpregu.00268.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Alexander BT, Rinewalt AN, Cockrell KL, Massey MB, Bennett WA, Granger JP. Endothelin type a receptor blockade attenuates the hypertension in response to chronic reductions in uterine perfusion pressure. Hypertension 37: 485–489, 2001. doi: 10.1161/01.hyp.37.2.485. [DOI] [PubMed] [Google Scholar]
  • 125. Yeung KR, Sunderland N, Lind JM, Heffernan S, Pears S, Xu B, Hennessy A, Makris A. Increased salt sensitivity in offspring of pregnancies complicated by experimental preeclampsia. Clin Exp Pharmacol Physiol 45: 1302–1308, 2018. doi: 10.1111/1440-1681.13008. [DOI] [PubMed] [Google Scholar]
  • 126. Morton JS, Levasseur J, Ganguly E, Quon A, Kirschenman R, Dyck JRB, Fraser GM, Davidge ST. Characterisation of the selective reduced uteroplacental perfusion (sRUPP) model of preeclampsia. Sci Rep 9: 9565, 2019. doi: 10.1038/s41598-019-45959-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Makris A, Thornton C, Thompson J, Thomson S, Martin R, Ogle R, Waugh R, McKenzie P, Kirwan P, Hennessy A. Uteroplacental ischemia results in proteinuric hypertension and elevated sFLT-1. Kidney Int 71: 977–984, 2007. doi: 10.1038/sj.ki.5002175. [DOI] [PubMed] [Google Scholar]
  • 128. Crews JK, Herrington JN, Granger JP, Khalil RA. Decreased endothelium-dependent vascular relaxation during reduction of uterine perfusion pressure in pregnant rat. Hypertension 35: 367–372, 2000. doi: 10.1161/01.hyp.35.1.367. [DOI] [PubMed] [Google Scholar]
  • 129. Bakrania BA, Hall ME, Shahul S, Granger JP. The reduced uterine perfusion pressure (RUPP) rat model of preeclampsia exhibits impaired systolic function and global longitudinal strain during pregnancy. Pregnancy Hypertens 18: 169–172, 2019. doi: 10.1016/j.preghy.2019.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Clayton AM, Shao Q, Paauw ND, Giambrone AB, Granger JP, Warrington JP. Postpartum increases in cerebral edema and inflammation in response to placental ischemia during pregnancy. Brain Behav Immun 70: 376–389, 2018. doi: 10.1016/j.bbi.2018.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Paauw ND, Joles JA, Spradley FT, Bakrania B, Zsengeller ZK, Franx A, Verhaar MC, Granger JP, Lely AT. Exposure to placental ischemia impairs postpartum maternal renal and cardiac function in rats. Am J Physiol Regul Integr Comp Physiol 312: R664–R670, 2017. doi: 10.1152/ajpregu.00510.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Booz GW, Kennedy D, Bowling M, Robinson T, Azubuike D, Fisher B, Brooks K, Chinthakuntla P, Hoang NH, Hosler JP, Cunningham MW Jr.. Angiotensin II type 1 receptor agonistic autoantibody blockade improves postpartum hypertension and cardiac mitochondrial function in rat model of preeclampsia. Biol Sex Differ 12: 58, 2021. doi: 10.1186/s13293-021-00396-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Brennan L, Morton JS, Quon A, Davidge ST. Postpartum vascular dysfunction in the reduced uteroplacental perfusion model of preeclampsia. PLoS One 11: e0162487, 2016. doi: 10.1371/journal.pone.0162487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Pulgar VM, Yamaleyeva LM, Varagic J, McGee C, Bader M, Dechend R, Brosnihan KB. Functional changes in the uterine artery precede the hypertensive phenotype in a transgenic model of hypertensive pregnancy. Am J Physiol Endocrinol Physiol 309: E811–E817, 2015. doi: 10.1152/ajpendo.00526.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Kräker K, O'Driscoll JM, Schütte T, Herse F, Patey O, Golic M, Geisberger S, Verlohren S, Birukov A, Heuser A, Müller DN, Thilaganathan B, Dechend R, Haase N. Statins reverse postpartum cardiovascular dysfunction in a rat model of preeclampsia. Hypertension 75: 202–210, 2020. doi: 10.1161/HYPERTENSIONAHA.119.13219. [DOI] [PubMed] [Google Scholar]
  • 136. Kedziora SM, Kräker K, Markó L, Binder J, Sugulle M, Gauster M, Müller DN, Dechend R, Haase N, Herse F. Kidney injury caused by preeclamptic pregnancy recovers postpartum in a transgenic rat model. Int J Mol Sci 22: 3762, 2021. doi: 10.3390/ijms22073762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Doridot L, Passet B, Méhats C, Rigourd V, Barbaux S, Ducat A, Mondon F, Vilotte M, Castille J, Breuiller-Fouché M, Daniel N, Le Provost F, Bauchet A-L, Baudrie V, Hertig A, Buffat C, Simeoni U, Germain G, Vilotte J-L, Vaiman D. Preeclampsia-like symptoms induced in mice by fetoplacental expression of STOX1 are reversed by aspirin treatment. Hypertension 61: 662–668, 2013. doi: 10.1161/HYPERTENSIONAHA.111.202994. [DOI] [PubMed] [Google Scholar]
  • 138. Ducat A, Doridot L, Calicchio R, Méhats C, Vilotte J-L, Castille J, Barbaux S, Couderc B, Jacques S, Letourneur F, Buffat C, Le Grand F, Laissue P, Miralles F, Vaiman D. Endothelial cell dysfunction and cardiac hypertrophy in the STOX1 model of preeclampsia. Sci Rep 6: 19196, 2016. doi: 10.1038/srep19196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Sutton EF, Gemmel M, Brands J, Gallaher MJ, Powers RW. Paternal deficiency of complement component C1q leads to a preeclampsia-like pregnancy in wild-type female mice and vascular adaptations postpartum. Am J Physiol Regul Integr Comp Physiol 318: R1047–R1057, 2020. doi: 10.1152/ajpregu.00353.2019. [DOI] [PubMed] [Google Scholar]
  • 140. Garrett N, Pombo J, Umpierrez M, Clark JE, Simmons M, Girardi G. Pravastatin therapy during preeclampsia prevents long-term adverse health effects in mice. JCI Insight 3: e120147, 2018. doi: 10.1172/jci.insight.120147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Stanley JL, Sulek K, Andersson IJ, Davidge ST, Kenny LC, Sibley CP, Mandal R, Wishart DS, Broadhurst DI, Baker PN. Sildenafil therapy normalizes the aberrant metabolomic profile in the Comt(−/−) mouse model of preeclampsia/fetal growth restriction. Sci Rep 5: 18241, 2015. doi: 10.1038/srep18241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Rueda-Clausen CF, Stanley JL, Thambiraj DF, Poudel R, Davidge ST, Baker PN. Effect of prenatal hypoxia in transgenic mouse models of preeclampsia and fetal growth restriction. Reprod Sci 21: 492–502, 2014. doi: 10.1177/1933719113503401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Kanasaki K, Palmsten K, Sugimoto H, Ahmad S, Hamano Y, Xie L, Parry S, Augustin HG, Gattone VH, Folkman J, Strauss JF, Kalluri R. Deficiency in catechol-O-methyltransferase and 2-methoxyoestradiol is associated with pre-eclampsia. Nature 453: 1117–1121, 2008. doi: 10.1038/nature06951. [DOI] [PubMed] [Google Scholar]
  • 144. Yamaleyeva LM, Pulgar VM, Lindsey SH, Yamane L, Varagic J, McGee C, daSilva M, Lopes Bonfa P, Gurley SB, Brosnihan KB. Uterine artery dysfunction in pregnant ACE2 knockout mice is associated with placental hypoxia and reduced umbilical blood flow velocity. Am J Physiol Endocrinol Metab 309: E84–E94, 2015. doi: 10.1152/ajpendo.00596.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Vieira Pimentel RL, Braga JF, Velloso EPP, Lautner RQ, de Oliveira ML, Todiras M, Alenina N, Bader M, de Sousa FG, Beier SL, Santos R. G-protein-coupled receptor MAS deletion produces a preeclampsia-like phenotype in FVB/N mice. Clin Sci (Lond) 137: 1249–1263, 2023. doi: 10.1042/CS20220819. [DOI] [PubMed] [Google Scholar]
  • 146. Lu F, Longo M, Tamayo E, Maner W, Al-Hendy A, Anderson GD, Hankins GDV, Saade GR. The effect of over-expression of sFlt-1 on blood pressure and the occurrence of other manifestations of preeclampsia in unrestrained conscious pregnant mice. Am J Obstet Gynecol 196: 396.e1–7, 2007. doi: 10.1016/j.ajog.2006.12.024. [DOI] [PubMed] [Google Scholar]
  • 147. Bytautiene E, Lu F, Tamayo EH, Hankins GDV, Longo M, Kublickiene K, Saade GR. Long-term maternal cardiovascular function in a mouse model of sFlt-1-induced preeclampsia. Am J Physiol Heart Circ Physiol 298: H189–H193, 2010. doi: 10.1152/ajpheart.00792.2009. [DOI] [PubMed] [Google Scholar]
  • 148. Murphy SR, LaMarca BB, Cockrell K, Granger JP. Role of endothelin in mediating soluble fms-like tyrosine kinase 1-induced hypertension in pregnant rats. Hypertension 55: 394–398, 2010. doi: 10.1161/HYPERTENSIONAHA.109.141473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Baijnath S, Soobryan N, Mackraj I, Gathiram P, Moodley J. The optimization of a chronic nitric oxide synthase (NOS) inhibition model of pre-eclampsia by evaluating physiological changes. Eur J Obstet Gynecol Reprod Biol 182: 71–75, 2014. doi: 10.1016/j.ejogrb.2014.08.021. [DOI] [PubMed] [Google Scholar]
  • 150. LaMarca BB, Cockrell K, Sullivan E, Bennett W, Granger JP. Role of endothelin in mediating tumor necrosis factor-induced hypertension in pregnant rats. Hypertension 46: 82–86, 2005. doi: 10.1161/01.HYP.0000169152.59854.36. [DOI] [PubMed] [Google Scholar]
  • 151. Warrington JP, Drummond HA, Granger JP, Ryan MJ. Placental ischemia-induced increases in brain water content and cerebrovascular permeability: role of TNF-α. Am J Physiol Regul Integr Comp Physiol 309: R1425–R1431, 2015. doi: 10.1152/ajpregu.00372.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Parrish MR, Ryan MJ, Glover P, Brewer J, Ray L, Dechend R, Martin JN Jr, Lamarca BB. Angiotensin II type 1 autoantibody induced hypertension during pregnancy is associated with renal endothelial dysfunction. Gend Med 8: 184–188, 2011. doi: 10.1016/j.genm.2011.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Wang H-P, Zhang W-H, Wang X-F, Zhu J, Zheng Y-Q, Xia Q, Zhi J-M. Exposure to AT1 receptor autoantibodies during pregnancy increases susceptibility of the maternal heart to postpartum ischemia-reperfusion injury in rats. Int J Mol Sci 15: 11495–11509, 2014. doi: 10.3390/ijms150711495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Bradshaw JL, Cushen SC, Ricci CA, Tucker SM, Gardner JJ, Little JT, Osikoya O, Goulopoulou S. Exposure to unmethylated CpG oligonucleotides disrupts blood pressure circadian rhythms and placental clock gene network in pregnant rats. Am J Physiol Heart Circ Physiol 325: H323–H337, 2023. doi: 10.1152/ajpheart.00154.2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Goulopoulou S, Wenceslau CF, McCarthy CG, Matsumoto T, Webb RC. Exposure to stimulatory CpG oligonucleotides during gestation induces maternal hypertension and excess vasoconstriction in pregnant rats. Am J Physiol Heart Circ Physiol 310: H1015–H1025, 2016. doi: 10.1152/ajpheart.00834.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. He B, Yang X, Li Y, Huang D, Xu X, Yang W, Dai Y, Zhang H, Chen Z, Cheng W. TLR9 (Toll-like receptor 9) agonist suppresses angiogenesis by differentially regulating vegfa (vascular endothelial growth factor A) and sFLT1 (soluble vascular endothelial growth factor receptor 1) in preeclampsia. Hypertension 71: 671–680, 2018. doi: 10.1161/HYPERTENSIONAHA.117.10510. [DOI] [PubMed] [Google Scholar]
  • 157. Tinsley JH, Chiasson VL, Mahajan A, Young KJ, Mitchell BM. Toll-like receptor 3 activation during pregnancy elicits preeclampsia-like symptoms in rats. Am J Hypertens 22: 1314–1319, 2009. doi: 10.1038/ajh.2009.185. [DOI] [PubMed] [Google Scholar]
  • 158. Sandgren JA, Deng G, Linggonegoro DW, Scroggins SM, Perschbacher KJ, Nair AR, Nishimura TE, Zhang SY, Agbor LN, Wu J, Keen HL, Naber MC, Pearson NA, Zimmerman KA, Weiss RM, Bowdler NC, Usachev YM, Santillan DA, Potthoff MJ, Pierce GL, Gibson-Corley KN, Sigmund CD, Santillan MK, Grobe JL. Arginine vasopressin infusion is sufficient to model clinical features of preeclampsia in mice. JCI Insight 3: e99403, 2018. doi: 10.1172/jci.insight.99403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Santillan MK, Santillan DA, Scroggins SM, Min JY, Sandgren JA, Pearson NA, Leslie KK, Hunter SK, Zamba GKD, Gibson-Corley KN, Grobe JL. Vasopressin in preeclampsia: a novel very early human pregnancy biomarker and clinically relevant mouse model. Hypertension 64: 852–859, 2014. [Erratum in Hypertension 65: e9, 2015]. doi: 10.1161/HYPERTENSIONAHA.114.03848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Xue P, Zheng M, Gong P, Lin C, Zhou J, Li Y, Shen L, Diao Z, Yan G, Sun H, Hu Y. Single administration of ultra-low-dose lipopolysaccharide in rat early pregnancy induces TLR4 activation in the placenta contributing to preeclampsia. PLoS One 10: e0124001, 2015. doi: 10.1371/journal.pone.0124001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Costantine MM, Tamayo E, Lu F, Bytautiene E, Longo M, Hankins GDV, Saade GR. Using pravastatin to improve the vascular reactivity in a mouse model of soluble fms-like tyrosine kinase-1-induced preeclampsia. Obstet Gynecol 116: 114–120, 2010. doi: 10.1097/AOG.0b013e3181e10ebd. [DOI] [PubMed] [Google Scholar]
  • 162. de Alwis N, Binder NK, Beard S, Mangwiro YT, Kadife E, Cuffe JS, Keenan E, Fato BR, Kaitu'u-Lino TJ, Brownfoot FC, Marshall SA, Hannan NJ. The L-NAME mouse model of preeclampsia and impact to long-term maternal cardiovascular health. Life Sci Alliance 5: e202201517, 2022. doi: 10.26508/lsa.202201517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Sáez T, Pageé A, Kirschenman R, Quon A, Spaans F, Davidge ST. A high cholesterol diet during late pregnancy impairs long-term maternal vascular function in mice. Arterioscler Thromb Vasc Biol 43: 120–132, 2023. doi: 10.1161/ATVBAHA.122.318421. [DOI] [PubMed] [Google Scholar]
  • 164. Sáez T, Spaans F, Kirschenman R, Sawamura T, Davidge ST. High-cholesterol diet during pregnancy induces maternal vascular dysfunction in mice: potential role for oxidized LDL-induced LOX-1 and AT1 receptor activation. Clin Sci (Lond) 134: 2295–2313, 2020. doi: 10.1042/CS20200764. [DOI] [PubMed] [Google Scholar]
  • 165. Johnson AC, Cipolla MJ. Impaired function of cerebral parenchymal arterioles in experimental preeclampsia. Microvasc Res 119: 64–72, 2018. doi: 10.1016/j.mvr.2018.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Cipolla MJ, Tremble SM, DeLance N, Johnson AC. Worsened stroke outcome in a model of preeclampsia is associated with poor collateral flow and oxidative stress. Stroke 54: 354–363, 2023. doi: 10.1161/STROKEAHA.122.041637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Cipolla MJ, Tremble S, DeLance N, Allison D, Johnson AC. Treatment with apocynin selectively restores hippocampal arteriole function and seizure-induced hyperemia in a model of preeclampsia. J Cereb Blood Flow Metab 42: 1425–1436, 2022. doi: 10.1177/0271678X221080092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Johnson AC, Tremble SM, Cipolla MJ. Experimental preeclampsia causes long-lasting hippocampal vascular dysfunction and memory impairment. Front Physiol 13: 889918, 2022. doi: 10.3389/fphys.2022.889918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. 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 39: 337–342, 2002. doi: 10.1161/hy02t2.102904. [DOI] [PubMed] [Google Scholar]
  • 170. Gillis EE, Mooney JN, Garrett MR, Granger JP, Sasser JM. Sildenafil treatment ameliorates the maternal syndrome of preeclampsia and rescues fetal growth in the Dahl salt-sensitive rat. Hypertension 67: 647–653, 2016. doi: 10.1161/HYPERTENSIONAHA.115.06071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Sones JL, Yarborough CC, O'Besso V, Lemenze A, Douglas NC. Genotypic analysis of the female BPH/5 mouse, a model of superimposed preeclampsia. PLoS One 16: e0253453, 2021. doi: 10.1371/journal.pone.0253453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Waker CA, Hwang AE, Bowman-Gibson S, Chandiramani CH, Linkous B, Stone ML, Keoni CI, Kaufman MR, Brown TL. Mouse models of preeclampsia with preexisting comorbidities. Front Physiol 14: 1137058, 2023. doi: 10.3389/fphys.2023.1137058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Turbeville HR, Taylor EB, Garrett MR, Didion SP, Ryan MJ, Sasser JM. Superimposed preeclampsia exacerbates postpartum renal injury despite lack of long-term blood pressure difference in the Dahl salt-sensitive rat. Hypertension 73: 650–658, 2019. doi: 10.1161/HYPERTENSIONAHA.118.12097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Maeda KJ, McClung DM, Showmaker KC, Warrington JP, Ryan MJ, Garrett MR, Sasser JM. Endothelial cell disruption drives increased blood-brain barrier permeability and cerebral edema in the Dahl SS/jr rat model of superimposed preeclampsia. Am J Physiol Heart Circ Physiol 320: H535–H548, 2021. doi: 10.1152/ajpheart.00383.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. 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. Biol Reprod 75: 899–907, 2006. doi: 10.1095/biolreprod.106.053603. [DOI] [PubMed] [Google Scholar]
  • 176. Arany Z, Hilfiker-Kleiner D, Karumanchi SA. Animal models of cardiovascular complications of pregnancy. Circ Res 130: 1763–1779, 2022. doi: 10.1161/CIRCRESAHA.122.320395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Gundling WE Jr, Wildman DE. A review of inter- and intraspecific variation in the eutherian placenta. Philos Trans R Soc Lond B Biol Sci 370: 20140072, 2015. doi: 10.1098/rstb.2014.0072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Roberts JM, Rich-Edwards JW, McElrath TF, Garmire L, Myatt L; Global Pregnancy Collaboration. Subtypes of preeclampsia: recognition and determining clinical usefulness. Hypertension 77: 1430–1441, 2021. doi: 10.1161/HYPERTENSIONAHA.120.14781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Hart BN, Shubrook JH, Mason T. Pregestational diabetes and family planning. Clin Diabetes 39: 323–328, 2021. doi: 10.2337/cd20-0062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Wang H, Li N, Chivese T, Werfalli M, Sun H, Yuen L, Hoegfeldt CA, Elise Powe C, Immanuel J, Karuranga S, Divakar H, Levitt N, Li C, Simmons D, Yang X; IDF Diabetes Atlas Committee Hyperglycaemia in Pregnancy Special Interest Group. IDF Diabetes Atlas: estimation of global and regional gestational diabetes mellitus prevalence for 2021 by International Association of Diabetes in Pregnancy Study Group’s Criteria. Diabetes Res Clin Pract 183: 109050, 2022. doi: 10.1016/j.diabres.2021.109050. [DOI] [PubMed] [Google Scholar]
  • 181. Oliveira AP, Calderon IMP, Costa RAA, Roscani MG, Magalhães CG, Borges VTM. Assessment of structural cardiac abnormalities and diastolic function in women with gestational diabetes mellitus. Diab Vasc Dis Res 12: 175–180, 2015. doi: 10.1177/1479164114563302. [DOI] [PubMed] [Google Scholar]
  • 182. Schannwell CM, Zimmermann T, Marx R, Plehn G, Leschke M, Strauer BE. [Alterations of left ventricular systolic and diastolic function in pregnant women with insulin-dependent diabetes mellitus]. Z Kardiol 91: 1024–1034, 2002. doi: 10.1007/s00392-002-0877-0. [DOI] [PubMed] [Google Scholar]
  • 183. Zawiejska A, Wender-Ożegowska E, Pietryga M, Brązert J. Maternal endothelial dysfunction and its association with abnormal fetal growth in diabetic pregnancy. Diabet Med 28: 692–698, 2011. doi: 10.1111/j.1464-5491.2011.03249.x. [DOI] [PubMed] [Google Scholar]
  • 184. Gutaj P, Wender-Ozegowska E. Diagnosis and management of IUGR in pregnancy complicated by type 1 diabetes mellitus. Curr Diab Rep 16: 39, 2016. doi: 10.1007/s11892-016-0732-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Jawerbaum A, White V. Animal models in diabetes and pregnancy. Endocr Rev 31: 680–701, 2010. doi: 10.1210/er.2009-0038. [DOI] [PubMed] [Google Scholar]
  • 186. Grupe K, Scherneck S. Mouse models of gestational diabetes mellitus and its subtypes: recent insights and pitfalls. Int J Mol Sci 24: 5982, 2023. doi: 10.3390/ijms24065982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Togashi Y, Shirakawa J, Okuyama T, Yamazaki S, Kyohara M, Miyazawa A, Suzuki T, Hamada M, Terauchi Y. Evaluation of the appropriateness of using glucometers for measuring the blood glucose levels in mice. Sci Rep 6: 25465, 2016. doi: 10.1038/srep25465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Damasceno DC, Netto AO, Iessi IL, Gallego FQ, Corvino SB, Dallaqua B, Sinzato YK, Bueno A, Calderon IMP, Rudge MVC. Streptozotocin-induced diabetes models: pathophysiological mechanisms and fetal outcomes. Biomed Res Int 2014: 819065, 2014. doi: 10.1155/2014/819065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Kottaisamy CPD, Raj DS, Prasanth Kumar V, Sankaran U. Experimental animal models for diabetes and its related complications—a review. Lab Anim Res 37: 23, 2021. doi: 10.1186/s42826-021-00101-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Burke SD, Dong H, Hazan AD, Croy BA. Aberrant endometrial features of pregnancy in diabetic NOD mice. Diabetes 56: 2919–2926, 2007. doi: 10.2337/db07-0773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Stanley JL, Cheung CC, Rueda-Clausen CF, Sankaralingam S, Baker PN, Davidge ST. Effect of gestational diabetes on maternal artery function. Reprod Sci 18: 342–352, 2011. doi: 10.1177/1933719110393029. [DOI] [PubMed] [Google Scholar]
  • 192. Stanley JL, Sankaralingam S, Baker PN, Davidge ST. Previous gestational diabetes impairs long-term endothelial function in a mouse model of complicated pregnancy. Am J Physiol Regul Integr Comp Physiol 299: R862–R870, 2010. doi: 10.1152/ajpregu.00200.2010. [DOI] [PubMed] [Google Scholar]
  • 193. Pasek RC, Gannon M. Advancements and challenges in generating accurate animal models of gestational diabetes mellitus. Am J Physiol Endocrinol Physiol 305: E1327–E1338, 2013. doi: 10.1152/ajpendo.00425.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. De Sousa RAL. Animal models of gestational diabetes: characteristics and consequences to the brain and behavior of the offspring. Metab Brain Dis 36: 199–204, 2021. doi: 10.1007/s11011-020-00661-9. [DOI] [PubMed] [Google Scholar]
  • 195. Aasa KL, Kwong KK, Adams MA, Croy BA. Analysis of maternal and fetal cardiovascular systems during hyperglycemic pregnancy in the nonobese diabetic mouse. Biol Reprod 88: 151, 2013. doi: 10.1095/biolreprod.112.105759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Lindegaard ML, Nielsen LB. Maternal diabetes causes coordinated down-regulation of genes involved with lipid metabolism in the murine fetal heart. Metabolism 57: 766–773, 2008. doi: 10.1016/j.metabol.2008.01.016. [DOI] [PubMed] [Google Scholar]
  • 197. Aryan L, Medzikovic L, Umar S, Eghbali M. Pregnancy-associated cardiac dysfunction and the regulatory role of microRNAs. Biol Sex Differ 11: 14, 2020. doi: 10.1186/s13293-020-00292-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Edupuganti MM, Ganga V. Acute myocardial infarction in pregnancy: current diagnosis and management approaches. Indian Heart J 71: 367–374, 2019. doi: 10.1016/j.ihj.2019.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Tweet MS, Lewey J, Smilowitz NR, Rose CH, Best PJM. Pregnancy-associated myocardial infarction: prevalence, causes, and interventional management. Circ Cardiovasc Interv. In press. doi: 10.1161/CIRCINTERVENTIONS.120.008687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Codsi E, Tweet MS, Rose CH, Arendt KW, Best PJM, Hayes SN. Spontaneous coronary artery dissection in pregnancy: what every obstetrician should know. Obstet Gynecol 128: 731–738, 2016. doi: 10.1097/AOG.0000000000001630. [DOI] [PubMed] [Google Scholar]
  • 201. De Viti D, Malvasi A, Busardò F, Beck R, Zaami S, Marinelli E. Cardiovascular outcomes in advanced maternal age delivering women. Clinical review and medico-legal issues. Medicina (Kaunas) 55: 658, 2019. doi: 10.3390/medicina55100658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Kealey A. Coronary artery disease and myocardial infarction in pregnancy: a review of epidemiology, diagnosis, and medical and surgical management. Can J Cardiol 26: 185–189, 2010. doi: 10.1016/s0828-282x(10)70397-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. James AH, Jamison MG, Biswas MS, Brancazio LR, Swamy GK, Myers ER. Acute myocardial infarction in pregnancy: a United States population-based study. Circulation 113: 1564–1571, 2006. doi: 10.1161/CIRCULATIONAHA.105.576751. [DOI] [PubMed] [Google Scholar]
  • 204. Roth A, Elkayam U. Acute myocardial infarction associated with pregnancy. J Am Coll Cardiol 52: 171–180, 2008. doi: 10.1016/j.jacc.2008.03.049. [DOI] [PubMed] [Google Scholar]
  • 205. Li J, Umar S, Iorga A, Youn J-Y, Wang Y, Regitz-Zagrosek V, Cai H, Eghbali M. Cardiac vulnerability to ischemia/reperfusion injury drastically increases in late pregnancy. Basic Res Cardiol 107: 271, 2012. doi: 10.1007/s00395-012-0271-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Li J, Ruffenach G, Kararigas G, Cunningham CM, Motayagheni N, Barakai N, Umar S, Regitz-Zagrosek V, Eghbali M. Intralipid protects the heart in late pregnancy against ischemia/reperfusion injury via Caveolin2/STAT3/GSK-3β pathway. J Mol Cell Cardiol 102: 108–116, 2017. doi: 10.1016/j.yjmcc.2016.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu X-Q, Zhang L. Gestational hypoxia and developmental plasticity. Physiol Rev 98: 1241–1334, 2018. doi: 10.1152/physrev.00043.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Tong W, Giussani DA. Preeclampsia link to gestational hypoxia. J Dev Orig Health Dis 10: 322–333, 2019. doi: 10.1017/S204017441900014X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Giussani DA, Davidge ST. Developmental programming of cardiovascular disease by prenatal hypoxia. J Dev Orig Health Dis 4: 328–337, 2013. doi: 10.1017/S204017441300010X. [DOI] [PubMed] [Google Scholar]
  • 210. West JB. Physiological effects of chronic hypoxia. N Engl J Med 376: 1965–1971, 2017. doi: 10.1056/NEJMra1612008. [DOI] [PubMed] [Google Scholar]
  • 211. Bailey BA, Donnelly M, Bol K, Moore LG, Julian CG. High altitude continues to reduce birth weights in Colorado. Matern Child Health J 23: 1573–1580, 2019. doi: 10.1007/s10995-019-02788-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Soria R, Julian CG, Vargas E, Moore LG, Giussani DA. Graduated effects of high-altitude hypoxia and highland ancestry on birth size. Pediatr Res 74: 633–638, 2013. doi: 10.1038/pr.2013.150. [DOI] [PubMed] [Google Scholar]
  • 213. Lichty JA, Ting RY, Bruns PD, Dyar E. Studies of babies born at high altitudes. I. Relation of altitude to birth weight. AMA J Dis Child 93: 666–669, 1957. doi: 10.1001/archpedi.1957.02060040668009. [DOI] [PubMed] [Google Scholar]
  • 214. Jensen GM, Moore LG. The effect of high altitude and other risk factors on birthweight: independent or interactive effects? Am J Public Health 87: 1003–1007, 1997. doi: 10.2105/ajph.87.6.1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Palmer SK, Moore LG, Young D, Cregger B, Berman JC, Zamudio S. Altered blood pressure course during normal pregnancy and increased preeclampsia at high altitude (3100 meters) in Colorado. Am J Obstet Gynecol 180: 1161–1168, 1999. doi: 10.1016/s0002-9378(99)70611-3. [DOI] [PubMed] [Google Scholar]
  • 216. Julian CG, Wilson MJ, Lopez M, Yamashiro H, Tellez W, Rodriguez A, Bigham AW, Shriver MD, Rodriguez C, Vargas E, Moore LG. Augmented uterine artery blood flow and oxygen delivery protect Andeans from altitude-associated reductions in fetal growth. Am J Physiol Regul Integr Comp Physiol 296: R1564–R1575, 2009. doi: 10.1152/ajpregu.90945.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Salinas CE, Patey OV, Murillo C, Gonzales M, Espinoza V, Mendoza S, Ruiz R, Vargas R, Perez Y, Montaño J, Toledo-Jaldin L, Badner A, Jimenez J, Peñaranda J, Romero C, Aguilar M, Riveros L, Arana I, Giussani DA. Preeclampsia and risk of maternal pulmonary hypertension at high altitude in Bolivia. J Dev Orig Health Dis 14: 523–531, 2023. doi: 10.1017/S2040174423000193. [DOI] [PubMed] [Google Scholar]
  • 218. Postigo L, Heredia G, Illsley NP, Torricos T, Dolan C, Echalar L, Tellez W, Maldonado I, Brimacombe M, Balanza E, Vargas E, Zamudio S. Where the O2 goes to: preservation of human fetal oxygen delivery and consumption at high altitude. J Physiol 587: 693–708, 2009. doi: 10.1113/jphysiol.2008.163634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Julian CG, Galan HL, Wilson MJ, Desilva W, Cioffi-Ragan D, Schwartz J, Moore LG. Lower uterine artery blood flow and higher endothelin relative to nitric oxide metabolite levels are associated with reductions in birth weight at high altitude. Am J Physiol Regul Integr Comp Physiol 295: R906–R915, 2008. doi: 10.1152/ajpregu.00164.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Kametas NA, McAuliffe F, Krampl E, Chambers J, Nicolaides KH. Maternal cardiac function during pregnancy at high altitude. BJOG 111: 1051–1058, 2004. doi: 10.1111/j.1471-0528.2004.00246.x. [DOI] [PubMed] [Google Scholar]
  • 221. Fallahi S, Houck JA, Euser AG, Julian CG, Moore LG, Lorca RA. High altitude differentially modulates potassium channel-evoked vasodilatation in pregnant human myometrial arteries. J Physiol 600: 5353–5364, 2022. doi: 10.1113/JP283741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Lorca RA, Lane SL, Bales ES, Nsier H, Yi HMi, Donnelly MA, Euser AG, Julian CG, Moore LG. High altitude reduces NO-dependent myometrial artery vasodilator response during pregnancy. Hypertension 73: 1319–1326, 2019. doi: 10.1161/HYPERTENSIONAHA.119.12641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Lorca RA, Matarazzo CJ, Bales ES, Houck JA, Orlicky DJ, Euser AG, Julian CG, Moore LG. AMPK activation in pregnant human myometrial arteries from high-altitude and intrauterine growth-restricted pregnancies. Am J Physiol Heart Circ Physiol 319: H203–H212, 2020. doi: 10.1152/ajpheart.00644.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Ong SS, Moore RJ, Warren AY, Crocker IP, Fulford J, Tyler DJ, Gowland PA, Baker PN. Myometrial and placental artery reactivity alone cannot explain reduced placental perfusion in pre-eclampsia and intrauterine growth restriction. BJOG 110: 909–915, 2003. [PubMed] [Google Scholar]
  • 225. Sweeney M, Wareing M, Mills TA, Baker PN, Taggart MJ. Characterisation of tone oscillations in placental and myometrial arteries from normal pregnancies and those complicated by pre-eclampsia and growth restriction. Placenta 29: 356–365, 2008. doi: 10.1016/j.placenta.2008.01.007. [DOI] [PubMed] [Google Scholar]
  • 226. Konje JC, Howarth ES, Kaufmann P, Taylor DJ. Longitudinal quantification of uterine artery blood volume flow changes during gestation in pregnancies complicated by intrauterine growth restriction. BJOG 110: 301–305, 2003. [PubMed] [Google Scholar]
  • 227. Lyall F, Robson SC, Bulmer JN. Spiral artery remodeling and trophoblast invasion in preeclampsia and fetal growth restriction: relationship to clinical outcome. Hypertension 62: 1046–1054, 2013. doi: 10.1161/HYPERTENSIONAHA.113.01892. [DOI] [PubMed] [Google Scholar]
  • 228. Mayhew TM. Changes in fetal capillaries during preplacental hypoxia: growth, shape remodelling and villous capillarization in placentae from high-altitude pregnancies. Placenta 24: 191–198, 2003. doi: 10.1053/plac.2002.0895. [DOI] [PubMed] [Google Scholar]
  • 229. van Patot MCT, Valdez M, Becky V, Cindrova-Davies T, Johns J, Zwerdling L, Jauniaux E, Burton GJ. Impact of pregnancy at high altitude on placental morphology in non-native women with and without preeclampsia. Placenta 30: 523–528, 2009. doi: 10.1016/j.placenta.2009.04.002. [DOI] [PubMed] [Google Scholar]
  • 230. Zamudio S. The placenta at high altitude. High Alt Med Biol 4: 171–191, 2003. doi: 10.1089/152702903322022785. [DOI] [PubMed] [Google Scholar]
  • 231. Lorca RA, Houck JA, Laurent LC, Matarazzo CJ, Baker K, Horii M, Nelson KK, Bales ES, Euser AG, Parast MM, Moore LG, Julian CG. High altitude regulates the expression of AMPK pathways in human placenta. Placenta 104: 267–276, 2021. doi: 10.1016/j.placenta.2021.01.010. [DOI] [PubMed] [Google Scholar]
  • 232. Zamudio S, Droma T, Norkyel KY, Acharya G, Zamudio JA, Niermeyer SN, Moore LG. Protection from intrauterine growth retardation in Tibetans at high altitude. Am J Phys Anthropol 91: 215–224, 1993. doi: 10.1002/ajpa.1330910207. [DOI] [PubMed] [Google Scholar]
  • 233. Herrera EA, Pulgar VM, Riquelme RA, Sanhueza EM, Reyes RV, Ebensperger G, Parer JT, Valdéz EA, Giussani DA, Blanco CE, Hanson MA, Llanos AJ. High-altitude chronic hypoxia during gestation and after birth modifies cardiovascular responses in newborn sheep. Am J Physiol Regul Integr Comp Physiol 292: R2234–R2240, 2007. doi: 10.1152/ajpregu.00909.2006. [DOI] [PubMed] [Google Scholar]
  • 234. Giussani DA, Salinas CE, Villena M, Blanco CE. The role of oxygen in prenatal growth: studies in the chick embryo. J Physiol 585: 911–917, 2007. doi: 10.1113/jphysiol.2007.141572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Zhou J, Xiao D, Hu Y, Wang Z, Paradis A, Mata-Greenwood E, Zhang L. Gestational hypoxia induces preeclampsia-like symptoms via heightened endothelin-1 signaling in pregnant rats. Hypertension 62: 599–607, 2013. doi: 10.1161/HYPERTENSIONAHA.113.01449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Higgins JS, Vaughan OR, Fernandez de Liger E, Fowden AL, Sferruzzi-Perri AN. Placental phenotype and resource allocation to fetal growth are modified by the timing and degree of hypoxia during mouse pregnancy. J Physiol 594: 1341–1356, 2016. doi: 10.1113/JP271057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Brain KL, Allison BJ, Niu Y, Cross CM, Itani N, Kane AD, Herrera EA, Giussani DA. Induction of controlled hypoxic pregnancy in large mammalian species. Physiol Rep 3: e12614, 2015. doi: 10.14814/phy2.12614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Turan S, Aberdeen GW, Thompson LP. Chronic hypoxia alters maternal uterine and fetal hemodynamics in the full-term pregnant guinea pig. Am J Physiol Regul Integr Comp Physiol 313: R330–R339, 2017. doi: 10.1152/ajpregu.00056.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Aljunaidy MM, Morton JS, Kirschenman R, Phillips T, Case CP, Cooke C-LM, Davidge ST. Maternal treatment with a placental-targeted antioxidant (MitoQ) impacts offspring cardiovascular function in a rat model of prenatal hypoxia. Pharmacol Res 134: 332–342, 2018. doi: 10.1016/j.phrs.2018.05.006. [DOI] [PubMed] [Google Scholar]
  • 240. Itani N, Skeffington KL, Beck C, Niu Y, Katzilieris-Petras G, Smith N, Giussani DA. Protective effects of pravastatin on the embryonic cardiovascular system during hypoxic development. FASEB J 34: 16504–16515, 2020. doi: 10.1096/fj.202001743R. [DOI] [PubMed] [Google Scholar]
  • 241. Skeffington KL, Beck C, Itani N, Niu Y, Shaw CJ, Giussani DA. Hypertension programmed in adult hens by isolated effects of developmental hypoxia in ovo. Hypertension 76: 533–544, 2020. doi: 10.1161/HYPERTENSIONAHA.120.15045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Aljunaidy MM, Morton JS, Cooke CL, Davidge ST. Maternal vascular responses to hypoxia in a rat model of intrauterine growth restriction. Am J Physiol Regul Integr Comp Physiol 311: R1068–R1075, 2016. doi: 10.1152/ajpregu.00119.2016. [DOI] [PubMed] [Google Scholar]
  • 243. Brain KL, Allison BJ, Niu Y, Cross CM, Itani N, Kane AD, Herrera EA, Skeffington KL, Botting KJ, Giussani DA. Intervention against hypertension in the next generation programmed by developmental hypoxia. PLoS Biol 17: e2006552, 2019. doi: 10.1371/journal.pbio.2006552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Mess A. The Guinea pig placenta: model of placental growth dynamics. Placenta 28: 812–815, 2007. doi: 10.1016/j.placenta.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 245. Hoes MF, Arany Z, Bauersachs J, Hilfiker-Kleiner D, Petrie MC, Sliwa K, van der Meer P. Pathophysiology and risk factors of peripartum cardiomyopathy. Nat Rev Cardiol 19: 555–565, 2022. doi: 10.1038/s41569-021-00664-8. [DOI] [PubMed] [Google Scholar]
  • 246. Hilfiker-Kleiner D, Kaminski K, Podewski E, Bonda T, Schaefer A, Sliwa K, Forster O, Quint A, Landmesser U, Doerries C, Luchtefeld M, Poli V, Schneider MD, Balligand J-L, Desjardins F, Ansari A, Struman I, Nguyen NQN, Zschemisch NH, Klein G, Heusch G, Schulz R, Hilfiker A, Drexler H. A cathepsin D-cleaved 16 kDa form of prolactin mediates postpartum cardiomyopathy. Cell 128: 589–600, 2007. doi: 10.1016/j.cell.2006.12.036. [DOI] [PubMed] [Google Scholar]
  • 247. Patten IS, Rana S, Shahul S, Rowe GC, Jang C, Liu L, Hacker MR, Rhee JS, Mitchell J, Mahmood F, Hess P, Farrell C, Koulisis N, Khankin EV, Burke SD, Tudorache I, Bauersachs J, del Monte F, Hilfiker-Kleiner D, Karumanchi SA, Arany Z. Cardiac angiogenic imbalance leads to peripartum cardiomyopathy. Nature 485: 333–338, 2012. doi: 10.1038/nature11040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Stapel B, Kohlhaas M, Ricke-Hoch M, Haghikia A, Erschow S, Knuuti J, Silvola JMU, Roivainen A, Saraste A, Nickel AG, Saar JA, Sieve I, Pietzsch S, Müller M, Bogeski I, Kappl R, Jauhiainen M, Thackeray JT, Scherr M, Bengel FM, Hagl C, Tudorache I, Bauersachs J, Maack C, Hilfiker-Kleiner D. Low STAT3 expression sensitizes to toxic effects of beta-adrenergic receptor stimulation in peripartum cardiomyopathy. Eur Heart J 38: 349–361, 2017. doi: 10.1093/eurheartj/ehw086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. van Spaendonck-Zwarts KY, Posafalvi A, van den Berg MP, Hilfiker-Kleiner D, Bollen IAE, Sliwa K, Alders M, Almomani R, van Langen IM, van der Meer P, Sinke RJ, van der Velden J, Van Veldhuisen DJ, van Tintelen JP, Jongbloed JDH. Titin gene mutations are common in families with both peripartum cardiomyopathy and dilated cardiomyopathy. Eur Heart J 35: 2165–2173, 2014. doi: 10.1093/eurheartj/ehu050. [DOI] [PubMed] [Google Scholar]
  • 250. Gramlich M, Michely B, Krohne C, Heuser A, Erdmann B, Klaassen S, Hudson B, Magarin M, Kirchner F, Todiras M, Granzier H, Labeit S, Thierfelder L, Gerull B. Stress-induced dilated cardiomyopathy in a knock-in mouse model mimicking human titin-based disease. J Mol Cell Cardiol 47: 352–358, 2009. doi: 10.1016/j.yjmcc.2009.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Ricke-Hoch M, Bultmann I, Stapel B, Condorelli G, Rinas U, Sliwa K, Scherr M, Hilfiker-Kleiner D. Opposing roles of Akt and STAT3 in the protection of the maternal heart from peripartum stress. Cardiovasc Res 101: 587–596, 2014. doi: 10.1093/cvr/cvu010. [DOI] [PubMed] [Google Scholar]
  • 252. Karamanlidis G, Lee CF, Garcia-Menendez L, Kolwicz SC Jr, Suthammarak W, Gong G, Sedensky MM, Morgan PG, Wang W, Tian R. Mitochondrial complex I deficiency increases protein acetylation and accelerates heart failure. Cell Metab 18: 239–250, 2013. doi: 10.1016/j.cmet.2013.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Otani K, Tokudome T, Kamiya CA, Mao Y, Nishimura H, Hasegawa T, Arai Y, Kaneko M, Shioi G, Ishida J, Fukamizu A, Osaki T, Nagai-Okatani C, Minamino N, Ensho T, Hino J, Murata S, Takegami M, Nishimura K, Kishimoto I, Miyazato M, Harada-Shiba M, Yoshimatsu J, Nakao K, Ikeda T, Kangawa K. Deficiency of cardiac natriuretic peptide signaling promotes peripartum cardiomyopathy-like remodeling in the mouse heart. Circulation 141: 571–588, 2020. doi: 10.1161/CIRCULATIONAHA.119.039761. [DOI] [PubMed] [Google Scholar]
  • 254. Zhu R-R, Chen Q, Liu Z-B, Ruan H-G, Wu Q-C, Zhou X-L. Inhibition of the Notch1 pathway induces peripartum cardiomyopathy. J Cell Mol Med 24: 7907–7914, 2020. doi: 10.1111/jcmm.15423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Lemmens K, Doggen K, De Keulenaer GW. Activation of the neuregulin/ErbB system during physiological ventricular remodeling in pregnancy. Am J Physiol Heart Circ Physiol 300: H931–H942, 2011. doi: 10.1152/ajpheart.00385.2010. [DOI] [PubMed] [Google Scholar]
  • 256. Shimizu T, Taguchi A, Higashijima Y, Kanki Y, Nakaki R, Urade Y, Wada Y. Inhibition of cardiac PERK signaling promotes peripartum cardiac dysfunction. Sci Rep 11: 18687, 2021. doi: 10.1038/s41598-021-98344-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Cooke CM, Davidge ST. Advanced maternal age and the impact on maternal and offspring cardiovascular health. Am J Physiol Heart Circ Physiol 317: H387–H394, 2019. doi: 10.1152/ajpheart.00045.2019. [DOI] [PubMed] [Google Scholar]
  • 258. Cleary-Goldman J, Malone FD, Vidaver J, Ball RH, Nyberg DA, Comstock CH, Saade GR, Eddleman KA, Klugman S, Dugoff L, Timor-Tritsch IE, Craigo SD, Carr SR, Wolfe HM, Bianchi DW, D'Alton M; FASTER Consortium. Impact of maternal age on obstetric outcome. Obstet Gynecol 105: 983–990, 2005. doi: 10.1097/01.AOG.0000158118.75532.51. [DOI] [PubMed] [Google Scholar]
  • 259. van Katwijk C, Peeters LL. Clinical aspects of pregnancy after the age of 35 years: a review of the literature. Hum Reprod Update 4: 185–194, 1998. doi: 10.1093/humupd/4.2.185. [DOI] [PubMed] [Google Scholar]
  • 260. Jacobsson B, Ladfors L, Milsom I. Advanced maternal age and adverse perinatal outcome. Obstet Gynecol 104: 727–733, 2004. doi: 10.1097/01.AOG.0000140682.63746.be. [DOI] [PubMed] [Google Scholar]
  • 261. Kenny LC, Lavender T, McNamee R, O'Neill SM, Mills T, Khashan AS. Advanced maternal age and adverse pregnancy outcome: evidence from a large contemporary cohort. PLoS One 8: e56583, 2013. doi: 10.1371/journal.pone.0056583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Cooke C-LM, Shah A, Kirschenman RD, Quon AL, Morton JS, Care AS, Davidge ST. Increased susceptibility to cardiovascular disease in offspring born from dams of advanced maternal age. J Physiol 596: 5807–5821, 2018. doi: 10.1113/JP275472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Shah A, Cooke C-LM, Kirschenman RD, Quon AL, Morton JS, Care AS, Davidge ST. Sex-specific effects of advanced maternal age on cardiovascular function in aged adult rat offspring. Am J Physiol Heart Circ Physiol 315: H1724–H1734, 2018. doi: 10.1152/ajpheart.00375.2018. [DOI] [PubMed] [Google Scholar]
  • 264. Morton JS, Care AS, Kirschenman R, Cooke CL, Davidge ST. Advanced maternal age worsens postpartum vascular function. Front Physiol 8: 465, 2017. doi: 10.3389/fphys.2017.00465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Velazquez MA, Smith CG, Smyth NR, Osmond C, Fleming TP. Advanced maternal age causes adverse programming of mouse blastocysts leading to altered growth and impaired cardiometabolic health in post-natal life. Hum Reprod 31: 1970–1980, 2016. doi: 10.1093/humrep/dew177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. El Assar M, Angulo J, Vallejo S, Peiró C, Sánchez-Ferrer CF, Rodríguez-Mañas L. Mechanisms involved in the aging-induced vascular dysfunction. Front Physiol 3: 132, 2012. doi: 10.3389/fphys.2012.00132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Nicholson CJ, Sweeney M, Robson SC, Taggart MJ. Estrogenic vascular effects are diminished by chronological aging. Sci Rep 7: 12153, 2017. doi: 10.1038/s41598-017-12153-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Pasha M, Wooldridge AL, Kirschenman R, Spaans F, Davidge ST, Cooke C-LM. Altered vascular adaptations to pregnancy in a rat model of advanced maternal age. Front Physiol 12: 718568, 2021. doi: 10.3389/fphys.2021.718568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Stanhewicz AE, Schwartz KS, Lee R. Increased stiffness of omental arteries from late pregnant women at advanced maternal age. Biosci Rep 43: BSR20231291, 2023. doi: 10.1042/BSR20231291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Wooldridge AL, Pasha M, Chitrakar P, Kirschenman R, Quon A, Spaans F, Sáez T, Cooke C-LM, Davidge ST. Advanced maternal age impairs uterine artery adaptations to pregnancy in rats. Int J Mol Sci 23: 9191, 2022. doi: 10.3390/ijms23169191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Care AS, Bourque SL, Morton JS, Hjartarson EP, Davidge ST. Effect of advanced maternal age on pregnancy outcomes and vascular function in the rat. Hypertension 65: 1324–1330, 2015. doi: 10.1161/HYPERTENSIONAHA.115.05167. [DOI] [PubMed] [Google Scholar]
  • 272. Lean SC, Heazell AEP, Dilworth MR, Mills TA, Jones RL. Placental dysfunction underlies increased risk of fetal growth restriction and stillbirth in advanced maternal age women. Sci Rep 7: 9677, 2017. doi: 10.1038/s41598-017-09814-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. van der Graaf AM, Zeeman GG, Groen H, Roberts C, Dekker GA. Non-invasive assessment of maternal hemodynamics in early pregnancy. Pregnancy Hypertens 3: 261–269, 2013. doi: 10.1016/j.preghy.2013.07.005. [DOI] [PubMed] [Google Scholar]
  • 274. Ling HZ, Garcia Jara P, Nicolaides KH, Kametas NA. Effect of maternal age on cardiac adaptation in pregnancy. Ultrasound Obstet Gynecol 58: 285–292, 2021. doi: 10.1002/uog.23614. [DOI] [PubMed] [Google Scholar]
  • 275. Jones TW, Houghton D, Cassidy S, MacGowan GA, Trenell MI, Jakovljevic DG. Bioreactance is a reliable method for estimating cardiac output at rest and during exercise. Br J Anaesth 115: 386–391, 2015. doi: 10.1093/bja/aeu560. [DOI] [PubMed] [Google Scholar]
  • 276. Whitten MK. Effect of exteroceptive factors on the oestrous cycle of mice. Nature 180: 1436, 1957. doi: 10.1038/1801436a0. [DOI] [PubMed] [Google Scholar]
  • 277. Caligioni CS. Assessing reproductive status/stages in mice. Curr Protoc Neurosci Appendix 4: Appendix 4I, 2009. doi: 10.1002/0471142301.nsa04is48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Byers SL, Wiles MV, Dunn SL, Taft RA. Mouse estrous cycle identification tool and images. PLoS One 7: e35538, 2012. doi: 10.1371/journal.pone.0035538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Heyne GW, Plisch EH, Melberg CG, Sandgren EP, Peter JA, Lipinski RJ. A simple and reliable method for early pregnancy detection in inbred mice. J Am Assoc Lab Anim Sci 54: 368–371, 2015. [PMC free article] [PubMed] [Google Scholar]
  • 280. Zhang J, Croy BA. Using ultrasonography to define fetal-maternal relationships: moving from humans to mice. Comp Med 59: 527–533, 2009. [PMC free article] [PubMed] [Google Scholar]
  • 281. Murray SA, Morgan JL, Kane C, Sharma Y, Heffner CS, Lake J, Donahue LR. Mouse gestation length is genetically determined. PLoS One 5: e12418, 2010. doi: 10.1371/journal.pone.0012418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Alexander BT, Kassab SE, Miller MT, Abram SR, Reckelhoff JF, Bennett WA, Granger JP. Reduced uterine perfusion pressure during pregnancy in the rat is associated with increases in arterial pressure and changes in renal nitric oxide. Hypertension 37: 1191–1195, 2001. doi: 10.1161/01.hyp.37.4.1191. [DOI] [PubMed] [Google Scholar]
  • 283. Bridges JP, Gilbert JS, Colson D, Gilbert SA, Dukes MP, Ryan MJ, Granger JP. Oxidative stress contributes to soluble fms-like tyrosine kinase-1 induced vascular dysfunction in pregnant rats. Am J Hypertens 22: 564–568, 2009. doi: 10.1038/ajh.2009.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Yallampalli C, Garfield RE. Inhibition of nitric oxide synthesis in rats during pregnancy produces signs similar to those of preeclampsia. Am J Obstet Gynecol 169: 1316–1320, 1993. doi: 10.1016/0002-9378(93)90299-x. [DOI] [PubMed] [Google Scholar]
  • 285. Lamarca B, Speed J, Ray LF, Cockrell K, Wallukat G, Dechend R, Granger J. Hypertension in response to IL-6 during pregnancy: role of AT1-receptor activation. Int J Interferon Cytokine Mediat Res 2011: 65–70, 2011. doi: 10.2147/IJICMR.S22329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Zenclussen AC, Fest S, Joachim R, Klapp BF, Arck PC. Introducing a mouse model for pre-eclampsia: adoptive transfer of activated Th1 cells leads to pre-eclampsia-like symptoms exclusively in pregnant mice. Eur J Immunol 34: 377–387, 2004. doi: 10.1002/eji.200324469. [DOI] [PubMed] [Google Scholar]
  • 287. Herrock O, Deer E, Amaral LM, Campbell N, Whitney D, Ingram N, Cornelius DC, Turner T, Hardy-Hardin J, Booz GW, Ibrahim T, LaMarca B. Inhibiting B cell activating factor attenuates preeclamptic symptoms in placental ischemic rats. Am J Reprod Immunol 89: e13693, 2023. doi: 10.1111/aji.13693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288. Collinot H, Marchiol C, Lagoutte I, Lager F, Siauve N, Autret G, Balvay D, Renault G, Salomon LJ, Vaiman D. Preeclampsia induced by STOX1 overexpression in mice induces intrauterine growth restriction, abnormal ultrasonography and BOLD MRI signatures. J Hypertens 36: 1399–1406, 2018. doi: 10.1097/HJH.0000000000001695. [DOI] [PubMed] [Google Scholar]
  • 289. Gillis EE, Williams JM, Garrett MR, Mooney JN, Sasser JM. The Dahl salt-sensitive rat is a spontaneous model of superimposed preeclampsia. Am J Physiol Regul Integr Comp Physiol 309: R62–R70, 2015. doi: 10.1152/ajpregu.00377.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290. Rhoads MK, Goleva SB, Beierwaltes WH, Osborn JL. Renal vascular and glomerular pathologies associated with spontaneous hypertension in the nonhuman primate Chlorocebus aethiops sabaeus. Am J Physiol Regul Integr Comp Physiol 313: R211–R218, 2017. doi: 10.1152/ajpregu.00026.2017. [DOI] [PubMed] [Google Scholar]
  • 291. Rebholz SL, Jones T, Burke KT, Jaeschke A, Tso P, D'Alessio DA, Woollett LA. Multiparity leads to obesity and inflammation in mothers and obesity in male offspring. Am J Physiol Endocrinol Metab 302: E449–E457, 2012. doi: 10.1152/ajpendo.00487.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Ogunmoroti O, Osibogun O, Kolade OB, Ying W, Sharma G, Vaidya D, Michos ED. Multiparity is associated with poorer cardiovascular health among women from the Multi-Ethnic Study of Atherosclerosis. Am J Obstet Gynecol 221: 631 e631–631.e16, 2019. doi: 10.1016/j.ajog.2019.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293. Jung JH, Lee GW, Lee JH, Byun MS, Yi D, Jeon SY, Jung GJ, Joung H, Shin SA, Kim YK, Kang KM, Sohn C-H, Lee DY. Multiparity, brain atrophy, and cognitive decline. Front Aging Neurosci 12: 159, 2020. doi: 10.3389/fnagi.2020.00159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294. Ritzel RM, Patel AR, Spychala M, Verma R, Crapser J, Koellhoffer EC, Schrecengost A, Jellison ER, Zhu L, Venna VR, McCullough LD. Multiparity improves outcomes after cerebral ischemia in female mice despite features of increased metabovascular risk. Proc Natl Acad Sci USA 114: E5673–E5682, 2017. doi: 10.1073/pnas.1607002114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Bonney EA. Demystifying animal models of adverse pregnancy outcomes: touching bench and bedside. Am J Reprod Immunol 69: 567–584, 2013. doi: 10.1111/aji.12102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Baylis C. The determinants of renal hemodynamics in pregnancy. Am J Kidney Dis 9: 260–264, 1987. doi: 10.1016/s0272-6386(87)80119-1. [DOI] [PubMed] [Google Scholar]
  • 297. Conrad KP. Possible mechanisms for changes in renal hemodynamics during pregnancy: studies from animal models. Am J Kidney Dis 9: 253–259, 1987. doi: 10.1016/s0272-6386(87)80118-x. [DOI] [PubMed] [Google Scholar]
  • 298. Novak J, Reckelhoff J, Bumgarner L, Cockrell K, Kassab S, Granger JP. Reduced sensitivity of the renal circulation to angiotensin II in pregnant rats. Hypertension 30: 580–584, 1997. doi: 10.1161/01.hyp.30.3.580. [DOI] [PubMed] [Google Scholar]
  • 299. Costantine MM. Physiologic and pharmacokinetic changes in pregnancy. Front Pharmacol 5: 65, 2014. doi: 10.3389/fphar.2014.00065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300. Morgan DJ. Drug disposition in mother and foetus. Clin Exp Pharmacol Physiol 24: 869–873, 1997. doi: 10.1111/j.1440-1681.1997.tb02707.x. [DOI] [PubMed] [Google Scholar]
  • 301. Liu XI, Momper JD, Rakhmanina N, van den Anker JN, Green DJ, Burckart GJ, Best BM, Mirochnick M, Capparelli EV, Dallmann A. Physiologically based pharmacokinetic models to predict maternal pharmacokinetics and fetal exposure to emtricitabine and acyclovir. J Clin Pharmacol 60: 240–255, 2020. doi: 10.1002/jcph.1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. Lee N, Hebert MF, Wagner DJ, Easterling TR, Liang CJ, Rice K, Wang J. Organic cation transporter 3 facilitates fetal exposure to metformin during pregnancy. Mol Pharmacol 94: 1125–1131, 2018. doi: 10.1124/mol.118.112482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303. Abduljalil K, Badhan RKS. Drug dosing during pregnancy-opportunities for physiologically based pharmacokinetic models. J Pharmacokinet Pharmacodyn 47: 319–340, 2020. doi: 10.1007/s10928-020-09698-w. [DOI] [PubMed] [Google Scholar]
  • 304. Tasnif Y, Morado J, Hebert MF. Pregnancy-related pharmacokinetic changes. Clin Pharmacol Ther 100: 53–62, 2016. doi: 10.1002/cpt.382. [DOI] [PubMed] [Google Scholar]
  • 305. Pariente G, Leibson T, Carls A, Adams-Webber T, Ito S, Koren G. Pregnancy-associated changes in pharmacokinetics: a systematic review. PLoS Med 13: e1002160, 2016. doi: 10.1371/journal.pmed.1002160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306. Horn CC, Kimball BA, Wang H, Kaus J, Dienel S, Nagy A, Gathright GR, Yates BJ, Andrews PLR. Why can’t rodents vomit? A comparative behavioral, anatomical, and physiological study. PLoS One 8: e60537, 2013. [Erratum in PLoS One 8, 2013]. doi: 10.1371/journal.pone.0060537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307. Tschiderer L, Seekircher L, Kunutsor SK, Peters SAE, O'Keeffe LM, Willeit P. Breastfeeding is associated with a reduced maternal cardiovascular risk: systematic review and meta-analysis involving data from 8 studies and 1,192,700 parous women. J Am Heart Assoc 11: e022746, 2022. doi: 10.1161/JAHA.121.022746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308. Scheiber IBR, Weiß BM, Kingma SA, Komdeur J. The importance of the altricial-precocial spectrum for social complexity in mammals and birds—a review. Front Zool 14: 3, 2017. doi: 10.1186/s12983-016-0185-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309. Rodriguez-Gonzalez GL, Bautista CJ, Rojas-Torres KI, Nathanielsz PW, Zambrano E. Importance of the lactation period in developmental programming in rodents. Nutr Rev 78: 32–47, 2020. doi: 10.1093/nutrit/nuaa041. [DOI] [PubMed] [Google Scholar]
  • 310. Freitas-de-Melo A, Orihuela A, Hötzel MJ, Ungerfeld R. What do we know and need to know about weaning in sheep? An overview of weaning practises, stress and welfare. Front Anim Sci 3: 823188, 2022. doi: 10.3389/fanim.2022.823188. [DOI] [Google Scholar]
  • 311. Suzette Tardif AC, Elmore D, Williams LE, Rice K. Reproduction and breeding of nonhuman primates. Nonhuman Primates in Biomedical Research 2012: 197–249, 2012. doi: 10.1016/B978-0-12-381365-7.00008-X. [DOI] [Google Scholar]
  • 312. Faust KM, Carouso-Peck S, Elson MR, Goldstein MH. The origins of social knowledge in altricial species. Annu Rev Dev Psychol 2: 225–246, 2020. doi: 10.1146/annurev-devpsych-051820-121446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. Pediatrics 150: e2022057988, 2022. doi: 10.1542/peds.2022-057988. [DOI] [PubMed] [Google Scholar]
  • 314. Gutkowska J, Jankowski M, Antunes-Rodrigues J. The role of oxytocin in cardiovascular regulation. Braz J Med Biol Res 47: 206–214, 2014. doi: 10.1590/1414-431X20133309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315. Uvnäs Moberg K, Ekström-Bergström A, Buckley S, Massarotti C, Pajalic Z, Luegmair K, Kotlowska A, Lengler L, Olza I, Grylka-Baeschlin S, Leahy-Warren P, Hadjigeorgiu E, Villarmea S, Dencker A. Maternal plasma levels of oxytocin during breastfeeding—a systematic review. PLoS One 15: e0235806, 2020. doi: 10.1371/journal.pone.0235806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316. Whitley J, Wouk K, Bauer AE, Grewen K, Gottfredson NC, Meltzer-Brody S, Propper C, Mills-Koonce R, Pearson B, Stuebe A. Oxytocin during breastfeeding and maternal mood symptoms. Psychoneuroendocrinology 113: 104581, 2020. doi: 10.1016/j.psyneuen.2019.104581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317. Hickman DL, Swan MP. Effects of age of pups and removal of existing litter on pup survival during cross-fostering between multiparous outbred mice. J Am Assoc Lab Anim Sci 50: 641–646, 2011. [PMC free article] [PubMed] [Google Scholar]
  • 318. Khan IY, Dekou V, Douglas G, Jensen R, Hanson MA, Poston L, Taylor PD. A high-fat diet during rat pregnancy or suckling induces cardiovascular dysfunction in adult offspring. Am J Physiol Regul Integr Comp Physiol 288: R127–R133, 2005. doi: 10.1152/ajpregu.00354.2004. [DOI] [PubMed] [Google Scholar]
  • 319. Fernandez-Twinn DS, Alfaradhi MZ, Martin-Gronert MS, Duque-Guimaraes DE, Piekarz A, Ferland-McCollough D, Bushell M, Ozanne SE. Downregulation of IRS-1 in adipose tissue of offspring of obese mice is programmed cell-autonomously through post-transcriptional mechanisms. Mol Metab 3: 325–333, 2014. doi: 10.1016/j.molmet.2014.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. McCurdy CE, Bishop JM, Williams SM, Grayson BE, Smith MS, Friedman JE, Grove KL. Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J Clin Invest 119: 323–335, 2009. doi: 10.1172/JCI32661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321. Mottola MF, Davenport MH, Ruchat S-M, Davies GA, Poitras VJ, Gray CE, Jaramillo Garcia A, Barrowman N, Adamo KB, Duggan M, Barakat R, Chilibeck P, Fleming K, Forte M, Korolnek J, Nagpal T, Slater LG, Stirling D, Zehr L. 2019 Canadian guideline for physical activity throughout pregnancy. Br J Sports Med 52: 1339–1346, 2018. doi: 10.1136/bjsports-2018-100056. [DOI] [PubMed] [Google Scholar]
  • 322. Hayman M, Brown WJ, Brinson A, Budzynski-Seymour E, Bruce T, Evenson KR. Public health guidelines for physical activity during pregnancy from around the world: a scoping review. Br J Sports Med 57: 940–947, 2023. doi: 10.1136/bjsports-2022-105777. [DOI] [PubMed] [Google Scholar]
  • 323. Davenport MH, Ruchat S-M, Poitras VJ, Jaramillo Garcia A, Gray CE, Barrowman N, Skow RJ, Meah VL, Riske L, Sobierajski F, James M, Kathol AJ, Nuspl M, Marchand A-A, Nagpal TS, Slater LG, Weeks A, Adamo KB, Davies GA, Barakat R, Mottola MF. Prenatal exercise for the prevention of gestational diabetes mellitus and hypertensive disorders of pregnancy: a systematic review and meta-analysis. Br J Sports Med 52: 1367–1375, 2018. doi: 10.1136/bjsports-2018-099355. [DOI] [PubMed] [Google Scholar]
  • 324. Davenport MH, McCurdy AP, Mottola MF, Skow RJ, Meah VL, Poitras VJ, Jaramillo Garcia A, Gray CE, Barrowman N, Riske L, Sobierajski F, James M, Nagpal T, Marchand A-A, Nuspl M, Slater LG, Barakat R, Adamo KB, Davies GA, Ruchat S-M. Impact of prenatal exercise on both prenatal and postnatal anxiety and depressive symptoms: a systematic review and meta-analysis. Br J Sports Med 52: 1376–1385, 2018. doi: 10.1136/bjsports-2018-099697. [DOI] [PubMed] [Google Scholar]
  • 325. Bull FC, Al-Ansari SS, Biddle S, Borodulin K, Buman MP, Cardon G, Carty C, Chaput JP, Chastin S, Chou R, Dempsey PC, DiPietro L, Ekelund U, Firth J, Friedenreich CM, Garcia L, Gichu M, Jago R, Katzmarzyk PT, Lambert E, Leitzmann M, Milton K, Ortega FB, Ranasinghe C, Stamatakis E, Tiedemann A, Troiano RP, van der Ploeg HP, Wari V, Willumsen JF. World Health Organization 2020 guidelines on physical activity and sedentary behaviour. Br J Sports Med 54: 1451–1462, 2020. doi: 10.1136/bjsports-2020-102955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326. McCurdy AP, Boulé NG, Sivak A, Davenport MH. Effects of exercise on mild-to-moderate depressive symptoms in the postpartum period: a meta-analysis. Obstet Gynecol 129: 1087–1097, 2017. doi: 10.1097/AOG.0000000000002053. [DOI] [PubMed] [Google Scholar]
  • 327. Cai C, Ruchat SM, Sivak A, Davenport MH. Prenatal exercise and cardiorespiratory health and fitness: a meta-analysis. Med Sci Sports Exerc 52: 1538–1548, 2020. doi: 10.1249/MSS.0000000000002279. [DOI] [PubMed] [Google Scholar]
  • 328. Skow RJ, King EC, Steinback CD, Davenport MH. The influence of prenatal exercise and pre-eclampsia on maternal vascular function. Clin Sci (Lond) 131: 2223–2240, 2017. doi: 10.1042/CS20171036. [DOI] [PubMed] [Google Scholar]
  • 329. Al-Huda F, Shapiro GD, Davenport MH, Bertagnolli M, Dayan N. Association between cardiorespiratory fitness and hypertensive disorders of pregnancy: a systematic review and meta-analysis. J Clin Med 11: 4364, 2022. doi: 10.3390/jcm11154364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330. Skow RJ, Davenport MH, Mottola MF, Davies GA, Poitras VJ, Gray CE, Jaramillo Garcia A, Barrowman N, Meah VL, Slater LG, Adamo KB, Barakat R, Ruchat S-M. Effects of prenatal exercise on fetal heart rate, umbilical and uterine blood flow: a systematic review and meta-analysis. Br J Sports Med 53: 124–133, 2019. doi: 10.1136/bjsports-2018-099822. [DOI] [PubMed] [Google Scholar]
  • 331. Wowdzia JB, Davenport MH. Cardiopulmonary exercise testing during pregnancy. Birth Defects Res 113: 248–264, 2021. doi: 10.1002/bdr2.1796. [DOI] [PubMed] [Google Scholar]
  • 332. Christopher SM, Donnelly G, Brockwell E, Bo K, Davenport MH, De Vivo M, Dufour S, Forner L, Mills H, Moore IS, Olson A, Deering RE. Clinical and exercise professional opinion of return-to-running readiness after childbirth: an international Delphi study and consensus statement. Br J Sports Med 58: 299–312, 2023. doi: 10.1136/bjsports-2023-107489. [DOI] [PubMed] [Google Scholar]
  • 333. Griffith JA, Garner KL, Bowdridge EC, DeVallance E, Schafner KJ, Engles KJ, Batchelor TP, Goldsmith WT, Wix K, Hussain S, Nurkiewicz TR. Nanomaterial inhalation during pregnancy alters systemic vascular function in a cyclooxygenase-dependent manner. Toxicol Sci 188: 219–233, 2022. doi: 10.1093/toxsci/kfac055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334. Garner KL, Bowdridge EC, Griffith JA, DeVallance E, Seman MG, Engels KJ, Groth CP, Goldsmith WT, Wix K, Batchelor TP, Nurkiewicz TR. Maternal nanomaterial inhalation exposure: critical gestational period in the uterine microcirculation is angiotensin II dependent. Cardiovasc Toxicol 22: 167–180, 2022. doi: 10.1007/s12012-021-09712-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335. Griffith JA, Dunn A, DeVallance E, Schafner KJ, Engles KJ, Batchelor TP, Goldsmith WT, Wix K, Hussain S, Bowdridge EC, Nurkiewicz TR. Maternal nano-titanium dioxide inhalation alters fetoplacental outcomes in a sexually dimorphic manner. Front Toxicol 5: 1096173, 2023. doi: 10.3389/ftox.2023.1096173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336. Bowdridge EC, DeVallance E, Garner KL, Griffith JA, Schafner K, Seaman M, Engels KJ, Wix K, Batchelor TP, Goldsmith WT, Hussain S, Nurkiewicz TR. Nano-titanium dioxide inhalation exposure during gestation drives redox dysregulation and vascular dysfunction across generations. Part Fibre Toxicol 19: 18, 2022. doi: 10.1186/s12989-022-00457-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337. Fournier SB, Kallontzi S, Fabris L, Love C, Stapleton PA. Effect of gestational age on maternofetal vascular function following single maternal engineered nanoparticle exposure. Cardiovasc Toxicol 19: 321–333, 2019. doi: 10.1007/s12012-019-09505-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338. Harris TR, Griffith JA, Clarke CEC, Garner KL, Bowdridge EC, DeVallance E, Engles KJ, Batchelor TP, Goldsmith WT, Wix K, Nurkiewicz TR, Rand AA. Distinct profiles of oxylipid mediators in liver, lung, and placenta after maternal nano-TiO(2) nanoparticle inhalation exposure. Env Sci Adv 2: 740–748, 2023. doi: 10.1039/d2va00300g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339. Kunovac A, Hathaway QA, Pinti MV, Goldsmith WT, Durr AJ, Fink GK, Nurkiewicz TR, Hollander JM. ROS promote epigenetic remodeling and cardiac dysfunction in offspring following maternal engineered nanomaterial (ENM) exposure. Part Fibre Toxicol 16: 24, 2019. doi: 10.1186/s12989-019-0310-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340. Hunter R, Baird B, Garcia M, Begay J, Goitom S, Lucas S, Herbert G, Scieszka D, Padilla J, Brayer K, Ottens AK, Suter MA, Barrozo ER, Hines C, Bleske B, Campen MJ. Gestational ozone inhalation elicits maternal cardiac dysfunction and transcriptional changes to placental pericytes and endothelial cells. Toxicol Sci 196: 238–249, 2023. doi: 10.1093/toxsci/kfad092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341. Miller CN, Dye JA, Ledbetter AD, Schladweiler MC, Richards JH, Snow SJ, Wood CE, Henriquez AR, Thompson LC, Farraj AK, Hazari MS, Kodavanti UP. Uterine artery flow and offspring growth in long-evans rats following maternal exposure to ozone during implantation. Environ Health Perspect 125: 127005, 2017. doi: 10.1289/EHP2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342. Weldy CS, Liu Y, Liggitt HD, Chin MT. In utero exposure to diesel exhaust air pollution promotes adverse intrauterine conditions, resulting in weight gain, altered blood pressure, and increased susceptibility to heart failure in adult mice. PLoS One 9: e88582, 2014. doi: 10.1371/journal.pone.0088582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343. Orzabal MR, Lunde-Young ER, Ramirez JI, Howe SYF, Naik VD, Lee J, Heaps CL, Threadgill DW, Ramadoss J. Chronic exposure to e-cig aerosols during early development causes vascular dysfunction and offspring growth deficits. Transl Res 207: 70–82, 2019. doi: 10.1016/j.trsl.2019.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.U.S. EPA. Guidelines for Exposure Assesssment. Washington, DC: U.S. Environmental Protection Agency, Risk Assessment Forum, 1992. [Google Scholar]
  • 345. Asgharian B, Price O, Borojeni AAT, Kuprat AP, Colby S, Singh RK, Gu W, Corley RA, Darquenne C. Influence of alveolar mixing and multiple breaths of aerosol intake on particle deposition in the human lungs. J Aerosol Sci 166: 106050, 2022. doi: 10.1016/j.jaerosci.2022.106050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346. Hussein T, Löndahl J, Paasonen P, Koivisto AJ, Petäjä T, Hämeri K, Kulmala M. Modeling regional deposited dose of submicron aerosol particles. Sci Total Environ 458–460: 140–149, 2013. doi: 10.1016/j.scitotenv.2013.04.022. [DOI] [PubMed] [Google Scholar]
  • 347. Kuprat AP, Jalali M, Jan T, Corley RA, Asgharian B, Price O, Singh RK, Colby S, Darquenne C. Efficient bi-directional coupling of 3D computational fluid-particle dynamics and 1D multiple path particle dosimetry lung models for multiscale modeling of aerosol dosimetry. J Aerosol Sci 151: 105647, 2021. doi: 10.1016/j.jaerosci.2020.105647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348. Rissler J, Gudmundsson A, Nicklasson H, Swietlicki E, Wollmer P, Löndahl J. Deposition efficiency of inhaled particles (15-5000 nm) related to breathing pattern and lung function: an experimental study in healthy children and adults. Part Fibre Toxicol 14: 10, 2017. doi: 10.1186/s12989-017-0190-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349. Ripplinger CM, Glukhov AV, Kay MW, Boukens BJ, Chiamvimonvat N, Delisle BP, Fabritz L, Hund TJ, Knollmann BC, Li N, Murray KT, Poelzing S, Quinn TA, Remme CA, Rentschler SL, Rose RA, Posnack NG. Guidelines for assessment of cardiac electrophysiology and arrhythmias in small animals. Am J Physiol Heart Circ Physiol 323: H1137–H1166, 2022. doi: 10.1152/ajpheart.00439.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350. Wenceslau CF, McCarthy CG, Earley S, England SK, Filosa JA, Goulopoulou S, Gutterman DD, Isakson BE, Kanagy NL, Martinez-Lemus LA, Sonkusare SK, Thakore P, Trask AJ, Watts SW, Webb RC. Guidelines for the measurement of vascular function and structure in isolated arteries and veins. Am J Physiol Heart Circ Physiol 321: H77–H111, 2021. doi: 10.1152/ajpheart.01021.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351. Mishra PK, Adameova A, Hill JA, Baines CP, Kang PM, Downey JM, Narula J, Takahashi M, Abbate A, Piristine HC, Kar S, Su S, Higa JK, Kawasaki NK, Matsui T. Guidelines for evaluating myocardial cell death. Am J Physiol Heart Circ Physiol 317: H891–H922, 2019. [Erratum in Am J Physiol Heart Circ Physiol 317: H1390, 2019]. doi: 10.1152/ajpheart.00259.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352. Brooks HL, Lindsey ML. Guidelines for authors and reviewers on antibody use in physiology studies. Am J Physiol Heart Circ Physiol 314: H724–H732, 2018. doi: 10.1152/ajpheart.00512.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353. Lindsey ML, Kassiri Z, Virag JAI, de Castro Brás LE, Scherrer-Crosbie M. Guidelines for measuring cardiac physiology in mice. Am J Physiol Heart Circ Physiol 314: H733–H752, 2018. doi: 10.1152/ajpheart.00339.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354. Lindsey ML, Bolli R, Canty JM Jr, Du X-J, Frangogiannis NG, Frantz S, Gourdie RG, Holmes JW, Jones SP, Kloner RA, Lefer DJ, Liao R, Murphy E, Ping P, Przyklenk K, Recchia FA, Schwartz Longacre L, Ripplinger CM, Van Eyk JE, Heusch G. Guidelines for experimental models of myocardial ischemia and infarction. Am J Physiol Heart Circ Physiol 314: H812–H838, 2018. doi: 10.1152/ajpheart.00335.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355. Lindsey ML, Gray GA, Wood SK, Curran-Everett D. Statistical considerations in reporting cardiovascular research. Am J Physiol Heart Circ Physiol 315: H303–H313, 2018. doi: 10.1152/ajpheart.00309.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Heart and Circulatory Physiology are provided here courtesy of American Physiological Society

RESOURCES