Open access peer-reviewed chapter

GDM-Induced Vascular Injury and Its Relationship with Fetal Metabolic Impairment

Written By

Cristian Espinoza

Submitted: 03 January 2022 Reviewed: 12 January 2022 Published: 17 February 2022

DOI: 10.5772/intechopen.102626

From the Edited Volume

Gestational Diabetes Mellitus - New Developments

Edited by Miroslav Radenković

Chapter metrics overview

601 Chapter Downloads

View Full Metrics

Abstract

Cardiovascular diseases are a significant health problem worldwide. To date, there is a lack of awareness that perinatal factors can predispose to CVD before birth. Gestational diabetes mellitus is an increasingly prevalent disease associated with poor fetal outcomes and CVD in the offspring. Evidence from the last decades suggests that GDM causes endothelial dysfunction and impairs nutrient transfer across the placenta to the fetus. These pathological features are associated with altered vascular and trophoblastic homeostasis in the placenta, predisposing the offspring to vascular injury, altered metabolic condition, and future CVD. This chapter focuses its discussion on the to-date understanding of GDM fetoplacental vascular and nutrient transfer impairment that causes, along with the latest advances, limitations, and questions that remain unresolved in this field.

Keywords

  • gestational diabetes mellitus
  • cardiovascular diseases
  • pregnancy
  • hyperglycemia
  • endothelial cells

1. Introduction

Almost one of every three adults worldwide dies because of cardiovascular diseases (CVD), making them the most prevalent cause of morbidity and mortality [1]. Several factors increase the risk of suffering a CVD. They can divide into two groups: modifiable and non-modifiable [2]. The former are those factors that can be controlled and modified by behavior, such as physical activity and diet, while the latter ones cannot be changed, like age and genetics. Environmental factors include air pollution and exposure to heavy metals, such as arsenic or lead, and the WHO recognizes them as important CVD risk factors. They could be considered “modifiable”; however, considering that most of the population affected by environmental pollution live in medium to low-income countries, their modification might be complex. An excellent review on this topic was published elsewhere [3]. The apparition of pathological conditions during pregnancy such as pre-eclampsia [4], maternal supraphysiological hypercholesterolemia (MSPH) [5], or gestational diabetes mellitus (GDM) [6] alters the fetal environment and is associated with an increase an increased risk of CVD in the offspring. They might be considered “between” modifiable and non-modifiable: In the gestational state, controlling the disease might prevent the fetal vascular impairment; however, after birth, there is a lack of evidence regarding treatments for improving their outcome and might be considered a non-modifiable factor. Most of the published research focuses on the repercussions of maternal health after suffering pregnancy disease [7, 8]. Still, their effects on the cardiovascular health of the fetus have been less described.

Over the last decades, the evidence associating pregnancy diseases and fetal outcomes has grown. Regarding MSPH, the apparition of fatty streaks on tunica intima of large arteries at fetal stages [9], probably related to alterations in nutrient transfer through the placenta [10], increases the risk of future cardiovascular events. Sadly, this condition is frequently underdiagnosed [11], and for a solid understanding of its prognosis more studies are needed. Preeclampsia is a relatively common complication of pregnancy [12]. It is associated with a slight but sustained increase in diastolic and systolic pressure on the offspring that seems to maintain for life [13, 14]. Preeclampsia also is related to Intrauterine Growth Restriction [15], which, in turn, is associated with an impaired vascular and metabolic condition [16], predisposing the offspring to worst cardiovascular outcomes. Finally, GDM is a more common disease with global prevalence between 6 and 7% (Europe and United States) and 9 and 13% (South and Central America, Asia, Africa) [17]. The prevalence over the last decades has been increasing in most countries [18, 19, 20, 21]; this might relate to the increase in maternal body mass index and the age of a pregnancy [18]. In terms of fetal cardiovascular impairment, a recent meta-analysis found that the offspring whose gestation was affected by GDM present higher basal glucose and systolic pressure [6]. In another large study, GDM pregnancies increased the prevalence of early-onset CVD by almost 30% in the offspring [22]. This worldwide statistical information urges researchers and clinicians to study the repercussions of GDM-complicated pregnancies further. Even more, an association between GDM and preeclampsia has been recently described [23, 24]. This relation can be explained at a systemic level by the increase in the age at which women become pregnant and the augment in body mass index told before; besides, both are related to damage on endothelial cells (EC), impairing vascular homeostasis [23]. Finally, between GDM and MSPH, a relation was recently suggested [25, 26], where EC in the placental vasculature and trophoblasts might have a crucial role; however, there is a considerable lack of evidence in this regard. At this point seems fair to suggest that EC is essential for the pathological development of the three conditions mentioned above. We will explore the GDM-induced vascular and trophoblastic injury and how it can probably impair fetal vascular health in the following pages.

1.1 Gestational diabetes mellitus pathophysiology

GDM is the apparition of spontaneous hyperglycemia in pregnancy without the previous diagnosis of a condition whose main feature is insulin resistance (IR) [27]. This definition is consistent with the evidence that GDM pathophysiology differs from pregnancies of women with prior diabetes in multiple aspects [28] as discussed later.

During a healthy pregnancy, the peripheral insulin sensitivity variates: In the early gestation increases to promote the fill up the glycogen and adipose stores [29], later it declines [30], increasing maternal systemic and placental glycemia. This reduction of insulin sensitivity (i.e., IR) occurs due to the pregnancy variation of systemic and placental hormones (for example, leptin, cortisol, estrogen, progesterone) [31] and is matched with a 2-fold increase in insulin secretion from pancreatic β-cells [32]. Late gestational hyperglycemia favors the transport of glucose to the fetus; however, it depletes the glycogen reservoirs and induces the use of fatty acids as fuel [33]. In GDM, maternal insulin sensitivity almost halves [34], implying two consequences: less accumulation of glycogen in both muscle and liver in the early pregnancy and faster use of them during late pregnancy. Furthermore, once the glycogen stores deplete, the use of fatty acids to obtain energy is more pronounced than in physiological pregnancy, leading to hypertriglyceridemia (HTG) (Figure 1).

Figure 1.

Metabolic differences between first and third trimester in healthy pregnancies, with a low degree of IR and previously diagnosed Diabetes mellitus. Previous low degree of insulin resistance increases the risk of developing GDM in the third trimester; however, since it courses without significant symptoms, previous IR repercussions are not usually assessed. PGDM alters the placenta’s formation in the first trimester, leading to more significant complications on the mother and the fetus in the third trimester.

Nonetheless, the reader is invited to reflect that a broad spectrum of clinical conditions related to the variable peripheral state of IR can exist [35, 36]. Finally, HTG and hyperglycemia alter placental vasculature [37] and fetal metabolic homeostasis [22, 38]. These features will be the focus of the following sections.

1.2 Gestational diabetes mellitus diagnose

Even when most of the pathophysiological features are known, reaching a diagnostic criterion for GDM has been troublesome. Huhn et al. [39] recently published a review of this topic. To date, one of the most widely accepted definitions is from the International Association of Diabetes and Pregnancy Study Group (IADPSG) [40]. The American Diabetes Association (ADA) agreed with IADPSG; however, first, they suggested a more flexible criterion than IADPS [41]. Table 1 summarizes both.

TestIADPSGADA
Fasting glucose≥92 mg/dL≥95 mg/dL
1-h glycemia after OGTT*≥180 mg/dL≥180 mg/dL
2-h glycemia after OGTT*≥153 mg/dL≥155 mg/dL

Table 1.

Diagnostic values for GDM.

OGTT: oral glucose tolerance test after a charge of 75 g of oral glucose.


• Both entities consider that this evaluation must be performed between 24 and 28 weeks of gestation.

• The first sample must be taken after 8 h of fasting.

The main difference between IADPS and ADA criteria is that the former considers that only one of the mentioned values needs to be altered to diagnose GDM. At the same time, ADA suggests that at least two of them must be present to diagnose GDM [41]. This slight discrepancy seems to be clinically significant: IADPS diagnostic of GDM increases two-fold [42] or three-fold [43] compared to ADA; moreover, using the IADPS criteria for diagnosis and treatment improves the adverse fetal outcomes of GDM [42, 43]. In this regard, ADA recent guidelines validated and included the IADPS criteria for GDM diagnosis [44].

1.3 Pregestational diabetes mellitus (PGDM)

Women’s pregestational condition has historically complexed the study of GDM. GDM tends to appear in women with a previous degree of IR, and insufficient insulin synthesis or release from the pancreas before gravidity [40]. However, for decades, GDM was described as “any degree of glucose intolerance with onset or first recognition during pregnancy” [42, 45], regardless of the prior existence of unrecognized IR. This definition implies a severe limitation. The test for GDM is usually performed between the second and third trimester; but the screening for metabolic perturbances on women at fertile age, before pregnancy, are not actively pursued or a worldwide practice. In this regard, at the time of the GDM diagnose, there are two potential scenarios (Figure 1):

  • GDM “de novo”: the increase of IR at the third trimester will trigger the disease, leading both mother and fetus to a trimester of hyperglycemia and HTG.

  • PGDM: the entire pregnancy will occur under a higher IR state, only being detected (and treated) from the third trimester and onwards.

Both conditions are clinically different. For example, birth weight over 4 kg, known as macrosomia, is associated with several fetal metabolic complications [46]. GDM is a risk factor of macrosomia; however, it has been recently suggested that PGDM might cause more severe and frequent metabolic complications, including macrosomia, in the fetus than GDM [28, 47]. A possible explanation for this might rely on more prolonged exposure to higher IR consequences, such as hyperglycemia and HTG. HTG in pregnancy on its own is associated with macrosomia [48]; besides, increased blood glucose, the primary manifestation of both GDM and PGDM, is also associated with poorer fetal outcomes [6, 49]. Both cause oxidative stress [50, 51], cytokine release, and meta-inflammation [52] in the forming placenta, impairing its ultrastructure and the nutrient transport to the fetus. Nonetheless, PGDM will expose the placental vasculature since its early formation to HTG and hyperglycemia. In the next sections, we will extensively discuss this topic.

1.4 Gestational diabetes mellitus induced vascular injury

As stated before, hyperglycemia and HTG are characteristic features of GDM and cause vascular injury on the placenta. Same as what happens on type 2 diabetes mellitus, GDM altered glucose metabolism on placental vasculature increases the production of reactive oxygen species (ROS) leading to oxidative stress (OS) [53, 54]. OS, in turn, favors the activation of Nuclear Factor kappa Beta and other pro-inflammatory pathways [55]. In GDM, the placenta itself expresses inflammatory cytokines [56]. Inflammation and OS will further induce systemic and placental IR, reducing the entry of glucose to cells [32, 57], impairing glycogen synthesis at the muscle and liver, leading to hyperglycemia and HTG. This oxidative and inflammatory state will also induce endothelial dysfunction (ED) impairing the vascular response to tissular metabolic needs, altering nutrient transfer to the fetus, and increasing the expression of adhesion molecules. To understand better the pathophysiological features of GDM on placental blood vessels and how it impairs the fetal metabolic condition, it is necessary first to summarize the main characteristics of the human placenta.

1.4.1 Development of the placental vascular system

In this section, we will explore the main features of placental development. For an in-depth study on this topic, the reader is invited to review the recent publication done by Turco et al. [58]. In brief, after the fertilization, the zygote will course with successive divisions forming the blastocyst, which will, in turn, adhere to the endometrium and invade it. The most external epithelial layer of the blastocyst will produce various trophoblast cell types and generate the primary syncytium below the implanted embryo [59]. The outer trophoblasts cells will differentiate and fusion, creating the syncytiotrophoblasts [60], whereas the inner cells will differentiate in cytotrophoblast. The syncytiotrophoblasts invade the endometrium and give rise to lacunas, spaces filled with maternal blood that will enlarge, merge, and develop the trabecular system of the forming placenta. The structure formed by both cell types around the lacunae is the primary villi. Later, the fetal mesenchyme will penetrate the villous core forming a structure known as secondary villi. Finally, vascular capillaries will appear within the center of fetal mesenchyme, forming the tertiary villi after the third gestation week. In the following weeks, angiogenesis predominates, increasing capillary density in the villi by developing new branches from preexisting vessels. Thus, the surface area for nutrient and oxygen exchange between the mother and the fetus increase [61]. At this point, in terms of vascular development, the placenta has reached its maturity (Figure 2). It is important to note that there are other essential structures in placentogenesis; however, they are beyond the scope of this chapter.

Figure 2.

The human term placenta. Maternal blood reaches the intervillous space (lacunae) through spiral arteries. Then, nutrients and oxygen cross the cytotrophoblasts from the microvillous membrane to the basal membrane and gets to the fetal blood vessels.

1.4.2 Diabetes impact on placental vascular formation

Tertiary villi arise at half of the second trimester. The pathological difference between PGDM and GDM becomes essential at this stage: from the implantation, and onwards, PGDM will expose the trophoblastic layer to an insulin-resistant, hyperglycemic, and hypoxic environment. Exploring the detrimental effects of both conditions is complicated since it needs the interruption of the pregnancy in human studies. Nonetheless, recent data permitted insight into the alterations caused by GDM or PGDM on the early placenta.

Spiral arteries in the endometrium are invaded by trophoblasts and remodeled [62]. This remodeling turns them into a resistance vessel, favoring the fell of arterial pressure, increasing placental blood flow. This process occurs by a coordinated proliferation, differentiation, and invasion of the trophoblasts, further forming the placenta. Several growth factors, including insulin-like growth factor I (IGF-I), and II (IGF-II) among others, released from the same trophoblasts [63] and other placental cell types, stimulate this process [64]. The placenta of GDM is heavier than healthy pregnancies, at least from the second trimester [65] and onwards [66]. This process is not fully understood; however, some findings have elucidated the role of growth factors. Placentas of IR pregnancies have an increased number of cytotrophoblasts, syncytiotrophoblasts, and EC due to a higher proliferation rate [64]. Consequently, placental vascularization in GDM is also enhanced by increased angiogenesis [67]. Differences in expression and secretion of growth factors from GDM trophoblasts themselves seem likely [68]. This increase in proliferation and angiogenesis has been shown in term placentas [68, 69]; yet, a recent study found that high IR is associated with a decrease in trophoblasts’ proliferation and increased apoptosis on first-trimester placentas [70]. Another recent work suggested that hyperinsulinemia can also exert those detrimental effects [71]. Apoptosis is low in early healthy pregnancies placentas [72], progressively increasing until term [73]. On GDM, apoptosis analysis has led to conflicting results, showing a decrease [74] or an increase [75] in term placentas. Different technical approaches or the criteria used to diagnose GDM might explain these discrepancies; therefore, more detailed studies are needed. In summary, IR impairs the signaling of growth factors on vascular and trophoblast cells, diminishing the development and invasion respectively at the first trimester; however, as gestation progresses, more growth factors are secreted in a compensatory manner further increasing the size, weight, and the number of blood vessels in the placenta.

Among growth factors, IGF-I and IGF-II are potent stimulators of placental vascular growth, acting through their cognate receptor or insulin receptor. It is important to note that the insulin receptor has two isoforms: A and B. Isoform B presents a sequence of 12 amino acids in the α subunit that A does not have. This slight difference gives them different intracellular signaling and substrate affinity. Isoform A is associated with a mitogenic phenotype via mitogen-activated protein kinases (MAPK), while Isoform B induces metabolic modulation via protein kinase B (Akt). Moreover, IGF-II interaction with insulin receptor A induces cell growth and invasion, while insulin activity on the same isoform protects from apoptosis [76]. This differential action may explain the differences observed in the regulation of apoptosis and cell cycle described above. Exposure to increased insulin levels reduces the insulin receptor and IGF-I receptor’s signaling via insulin response element I and downstream targets such as Akt [63]. At this point seems fair to hypothesize that IR impairs placental vascular development by altering the insulin and IGF-I receptor signaling, dysregulating proliferation, and apoptosis. This impairment might explain the high immaturity level of the villous observed in the GDM placenta [77]. Concordant with this hypothesis, human umbilical veins endothelial cells (HUVEC) increase MAPK signaling probably via isoform A of the insulin receptor in GDM [78]. Insulin exposure reestablishes the downstream signaling and membrane expression of both isoforms [78], making it an attractive therapeutic alternative; however, the effectiveness of insulin is highly dependent on the previous IR state. Indeed, obese women that develop GDM respond worse to insulin treatment than lean, diminishing the insulin receptor presence at the membrane and lesser downstream signaling [79, 80]. It is important to note that maternal obesity does not mean necessary IR; however, since most studies do not present evidence from the pregestational state, this suggestion seems fair to be made. More studies are needed taking this consideration since insulin does not seem to be always the better option. An excellent review on this matter has been published elsewhere [81].

Finally, disruption of insulin and IGF receptors signaling, observed in IR states, is related to insufficient trophoblasts invasion, pregnancy-associated hypertension, and increased pregnancy complications, including abortion [63]. In this regard, insulin signaling in the placenta seems crucial and will focus on in the next section.

1.4.3 Placental vasomotor alterations on GDM

The human placenta has no autonomic innervation, so vascular tone regulation is performed by the myogenic tone and humoral and metabolic factors. Humoral factors include norepinephrine [82], renin-angiotensin system (RAS), and vasopressin [83]. The three of them impair invasion of the trophoblast in spiral arteries and alter placental vascular homeostasis. This phenomenon has been studied in pre-eclampsia; however, in GDM, there is a lack of evidence pointing to its potential pathological role. Strikingly, GDM increases the risk of pre-eclampsia from the first trimester and onwards [84]. Indeed, GDM curses with some of the same pre-eclampsia’s placental vascular complications (i.e., placental hypoxia and ED) [85]. In particular, maternal vasopressin does not seem to affect fetal blood flow [86], same as norepinephrine [87]. Nonetheless, the latter is related to a reduction in fetal oxygen delivery. This is likely to happen in pregnancies of women with prior diabetes [88] and GDM [89]: both conditions increase catecholamines plasmatic concentration in part because of hyperglycemia [90]. Moreover, norepinephrine augments IR [91, 92], and epinephrine diminishes insulin secretion from the pancreas [93]. In summary, upregulation of catecholamines in GDM negatively impacts placental vessel homeostasis; however, further studies are needed to explore this issue.

Several studies have highlighted the physiological role of the RAS system in placental development and function. A review in this regard has been recently published elsewhere [94]. In brief, the placenta presents all the components of RAS [95]. After implantation, tissular hypoxia induces syncytiotrophoblast formation, the remodeling of the spiral arteries, and angiogenesis. Angiotensin II receptor 1 (AT1R) expression is increased by hypoxia in trophoblasts and spiral arteries, augmenting the expression of angiogenic factors [96]. In healthy pregnancies, AT1R is highly expressed in the trophoblasts in the first and second trimester, declining its levels on the third [97]. However, if hypoxia persists, the expression of AT1R remains high until the end of the pregnancy [98]. As mentioned above, GDM incurs placental hypoxia, which might increase AT1R expression in trophoblasts [99], vascularity in the placenta and placental weight. AT1R expression due to GDM also increases in other vascular beds in rodent models, increasing vascular resistance and systemic arterial pressure [99]. Further, GDM increases the plasma concentration of angiotensin II (AGII), and permanent exposure to AGII induces vasoconstriction, diminishing placental blood flow and fetal oxygen delivery [100]. Also, IR in GDM may cause hyperinsulinemia, which in turn enhances the AT1R [101] and AGII [102] expression. In this regard, the relation between RAS and GDM seems to be even more complex. Higher plasma levels of soluble renin/prorenin receptor in the early pregnancy relate to an increased risk of developing GDM in late pregnancy [103]. This observation is in concordance with the fact that inhibitors of RAS, such as losartan, improve the vascular condition in human diabetes [104, 105] and rodent models of GDM [106]. Furthermore, GDM also increases the plasma concentration of aldosterone [107], an end product of RAS. Interestingly, hyperaldosteronism is associated with ED [108], which will be the subject of the following section. Nevertheless, to the best of my knowledge, this issue has not been assessed on fetoplacental vessels of GDM. Finally, increased AGII umbilical cord levels are associated with increased IR in GDM offspring [109]. Lesser perfusion of the β-cells can explain this due to vasoconstriction and a reduction of insulin sensitivity [110]. Indeed, blockade of RAS ameliorates IR [111]. Both processes converge in EC, where AGII increases ROS production, favoring oxidative stress (OS) [112]. In turn, GDM placenta incurs in OS [54], which impairs insulin signaling in multiple points and induces an inflammatory response mediated by Nuclear Factor kappa B, JNK, and p38 MAPK [113]. On the other hand, AT1R stimulation increases the apoptosis in villous explants and trophoblasts, which associates with pre-eclampsia [114], an event that might also happen in GDM; however, further studies are needed to explore this intricate process.

1.4.4 Endothelial dysfunction on GDM

ED is characterized by imbalanced vasodilation and vasoconstriction, elevated ROS, inflammation, and a deficit of nitric oxide (NO) bioavailability [115, 116]. All these phenomena occur in the GDM placenta, leading to an increased vascular tone and reduced perfusion.

Arachidonic acid is the precursor of thromboxane A2 (TXA2), a vasoconstrictor, and prostacyclin, a vasodilator. The synthesis of both can occur in EC. TXA2 acts through the TXA2 receptor (TR), present in the human umbilical vein. Besides, non-enzymatic oxidation of arachidonic acid produces isoprostanes [117], which can also interact with TR and induce constriction. GDM placentas show an increased synthesis of isoprostanes [54], probably due to the increased production of ROS. In GDM [118] and preeclampsia [119] the prostacyclin/TXA2 ratio is lower in the placenta. Interestingly, OS in trophoblasts increases the concentration of TXA2 but not prostacyclin, pointing to ROS as the responsible for this mechanism that increases the vascular tone. Endothelium-derived hyperpolarization (EDH) is mostly unexplored in placental vessels, yet it may play a role in GDM placental vascular impairment. EDH exerts vasodilation via stimulation of the Ca2+-activated K+ channels, which hyperpolarize vascular smooth muscle cells (VSMC) [120]. It is hard to guess if GDM alters this mechanism. Preeclamptic pregnancies show a lesser EDH effect [121]; however, type 2 diabetes mellitus increases the EDH effect [122]. Further studies are needed to elucidate if EDH impairs or compensates ED in GDM.

Nitric oxide (NO) is probably the most characterized endothelium-derived vasodilator agent. Indeed, some consider that NO is the most potent vasodilator in the human placenta [123]. Due to its biological relevance, it is not surprising that its bioavailability is highly regulated. For instance, NO depends on the cellular intake of L-Arginine and the activity of the nitric oxide synthases. In EC, endothelial nitric oxide synthase (eNOS) is the primary source of NO, and cytosolic calcium, protein kinase A, and AKT favor the activity of this enzyme [124]. Insulin stimulates eNOS via AKT; besides, diabetes impairs this stimulation reducing eNOS activity, while reduction of NO induces IR, forming a vicious cycle [125]. On the other hand, NO acts on the VSMC, causing dilation via guanylyl-cyclase; however, it favors apoptosis [126] and inhibits proliferation [127] of the same cell type. VSMC apoptosis reduces the capability of resistance vessels to contract; in contrast, AGII favors proliferation via ROS activation of p38 MAPK [128]. Interestingly, a recent publication observed that GDM increases the insulin receptor isoform A and IGF 1R [129]. This gives consistency to the observations stated before: GDM enhances RAS and insulin receptor isoform A signaling in the placenta, both favoring the proliferation of VSMC; however, even when the machinery to produce NO upregulates in GDM [130], a reduction in its bioavailability is observed probably due to depletion by oxidative stress [131]. In turn, NO reduction inhibits apoptosis and further favors proliferation of VSMC, which will increase vascular tone, reduce perfusion, increasing hypoxia, and stimulate angiogenesis and even more OS. Nonetheless, even when consistent, this idea (Figure 3) needs further experimental support.

Figure 3.

Mechanisms of GDM-induced endothelial dysfunction in the human placenta. Insulin resistance, hyperinsulinemia, hyperglycemia, hypertriglyceridemia (HTG), and high plasma concentration of free fatty acids (FFA) characterize GDM. These alterations induce vasoconstriction, hypoxia, and reactive oxygen species (ROS) production. ROS, in turn, will increase thromboxane A2 (TxA2) and isoprostanes in endothelial cells, further favoring vasoconstriction. ROS also upregulates the adenosine signaling, and the adenosine/L-Arginine/nitric oxide axis will be upregulated; however, insulin resistance diminishes endothelial nitric oxide synthase (eNOS) phosphorylation and nitric oxide (NO) production. Also, ROS will interact with NO and produce peroxynitrite, reducing NO bioavailability.

l-Arginine also determines the synthesis of NO by eNOS. l-Arginine is transformed in l-citrulline for NO production by eNOS [123]; so, NO production is dependent on intracellular l-Arginine content. Cationic amino acid transporter 1 (hCAT-1) is the main responsible for the entry of l-Arginine to the cell in the human [132]. Interestingly, insulin, OS and the activation of adenosine receptor A 2A (ARA2A) induce hCAT-1 expression [133]. In this regard, even when in GDM impairs insulin signaling, OS and the activation of ARA2A will favor the expression of hCAT-1 and secure the l-Arginine entry. OS can also induce the activation of adenosine receptor [134]. However, the insulin effect over hCAT-1 expression and activity has been described as requiring functional ARA2A in HUVEC [135]. GDM hinders adenosine transport to the cell, increasing its extracellular concentration [136, 137]. Extracellular adenosine will activate ARA2A, which will induce vasodilation [133]. Interestingly, adenosine can also interact with adenosine receptor A 2B (ARA2B), which is expressed in microvascular EC and induces angiogenesis [138]. The high adenosine concentration facilitates the ARA2B activation and may relate to the increased vascularization and weight observed in GDM placentas. A recent work has shown that adenosine induces fetal vessels constriction; however, GDM impairs its vasoconstrictor effect [139]. Going back to the above, even when the whole adenosine/l-Arginine/NO axis raises in GDM, the lesser bioavailability of NO impedes its biological effect. Likewise, NO deficit might explain why GDM reduces the insulin vasodilatory effect [131]. In this regard, endothelial dysfunction by GDM not only affects vasodilation but vasoconstriction as well, hindering the capability of the endothelium to regulate the vascular tone.

Finally, it is crucial to note that hypoxia [140] and hyperglycemia [141] induce OS. Interestingly, hyperglycemia on its own can induce hypoxia [142]. Mitochondrial impairment is probably the most important source of ROS in GDM; an excellent review has been made elsewhere [143]. Further, a recent work described mitochondrial dysfunction in cytotrophoblast and syncytiotrophoblast from GDM pregnancies. The latter seems to be more comprised in terms of ATP generation and increases the expression of antioxidants [144]. Nonetheless, this impairment is more profound when higher grades of IR are present [113] and the pregestational condition is highly relevant [143]. Insulin can increase the production of antioxidants; however, in GDM placentas, the expression of antioxidants is increased constantly [145], making them less responsive to future oxidants insults. Finally, ROS can react non-enzymatically with NO, producing peroxynitrite, which has been shown to inhibit mitochondrial respiration and damaged mitochondria [146], making a vicious cycle for ROS production.

1.4.5 Placental altered nutrient transfer on GDM and fetal metabolic injury

Hyperglycemia and HTG are the most common metabolic alterations in GDM. Recent work evidenced that HTG in early pregnancy is related to IR, β-cell dysfunction, and hyperglycemia [147]. Umbilical cord blood analysis has demonstrated that GDM causes fetal hyperinsulinemia proportional to maternal IR [148]. Triglyceridemia remains unaltered, but LDL concentrations increase and HDL diminishes in cord blood of GDM deliveries and directly associates with macrosomia [149]. Intriguingly, triglyceridemia remains unaltered since maternal HTG is better related to macrosomia than hyperglycemia itself [150, 151, 152]. In this regard, the relationship observed between HTG and macrosomia in GDM might have two possible causes: an increase in the fetal delivery of free fatty acids (FFA) posterior to the action of lipases or the impairment of placental homeostasis due to ED. The first hypothesis does not seem likely: even when GDM curses with high FFA maternal plasma concentration [153], cord blood FFA content remains unaltered in GDM deliveries [154]. In this regard, the second hypothesis seems more plausible. HTG is related to ED; however, a mechanistic explanation is lacking to date. A recent review was made about this topic elsewhere [155]. A probable explanation for ED lies in macrophage activation by triglyceride-rich lipoproteins like Very-Low-Density Lipoprotein (VLDL) [156]. VLDL also induces ROS production and expression of inflammation mediators such as Tumoral Necrosis Factor α (TNF-α) in EC [157]. Interestingly, TNF-α favors IR and hyperinsulinemia in GDM [32], hindering insulin-mediated vasodilation. Also, the oxidative environment induced by triglycerides may favor the NO consumption, establishing the ED. Nonetheless, further studies are needed to address this issue in GDM placentas.

Finally, glucose transport in the placenta is regulated by maternal glycemia and by the expression and activity of glucose transporters (GLUT). For transportation from the mother to the fetus, glucose must go through the microvillous membrane (MVM), at the maternal side, to the basal membrane (BM) on the fetal side [158]. At least 6 GLUT transporters have been identified in the placenta: GLUT1, GLUT3, GLUT4, GLUT8, GLUT9, and GLUT12. Nonetheless, the most abundant isoforms in the placenta are GLUT1 and GLUT4. GLUT1 levels increase in syncytiotrophoblasts along with the pregnancy progression [159]. GLUT1 expresses in the MVM 3-fold than in the BM. Thus, crossing the BM is the rate-limiting step for glucose transport to fetal circulation [160]. Indeed, increased content of GLUT1 is correlated proportionally with fetal weight and macrosomia [161]. GLUT4 expression, contrarily to what was thought before [159], increases during gestation in the MVM, but only in healthy lean women [162]. In GDM, interestingly, insulin lowers mRNA of GLUT4; besides, various authors found increased GLUT1 expression [79, 163, 164]. Even more, GLUT1 upregulation is more profound in PGDM [164]. In this regard, it seems fair to suggest that GLUT 1 in the BM is critical for GDM pregnancy complications due to increased glucose transport [165]. Hyperglycemia should limit GLUT1 expression in trophoblasts and favor its movement from the membrane to the cytoplasm [166, 167]; however, in GDM, this does not seem to happen. A mechanistic study is necessary to address this issue. Finally, an increased transfer of maternal insulin to the fetus could explain hyperinsulinemia observed in the fetal cord of GDM deliveries. Nonetheless, near 1% maternal insulin crosses the placenta [168]. This could hardly cause an increase in fetal insulinemia; however, it may contribute. In this regard, the Modified Pedersen hypothesis offers a better explanation: Maternal hyperglycemia passes through the placenta to the fetus; then, from the second trimester and onwards, the fetal pancreas responds to hyperglycemia with hyperinsulinemia, further favoring glucose disposition in fat stores and the anabolic effects of insulin, resulting in macrosomia [169]. This could also explain the vascular alterations observed in the GDM offspring; however, further research for addressing this issue is needed.

Advertisement

2. Conclusion

GDM is a complex condition that affects both fetus and mother. Its impact on the offspring includes vascular and metabolic impairment before birth, predisposing them to early CVD. The real prevalence of GDM worldwide is unknown and might go beyond our expectations since it is mostly underdiagnosed. Moreover, the differential impact of previously diagnosed diabetes in pregnancy has begun to elucidate in the last few decades. On the other hand, the reader is invited to reflect that the pathological IR state in pregnancy is not a “black-or-white” matter but a continuous spectrum of possible conditions and fetal outcomes that needs to be assessed in every pregnancy individually. Including the assessment of HbA1c and lipid profile test in the first trimester, evaluation might improve the diagnosis of PGDM and foresee the future GDM development.

Previous IR state and PGDM hinder syncytiotrophoblast invasion in maternal vessels and the placenta formation; however, there is still much to research and learn from this subject. After development, GDM will continuously expose the placenta to a hypoxic environment that will impair vascular function due to increased OS and inflammation. HTG, hyperglycemia, and increased FFA will favor this pro-oxidant environment, causing ED. The regulation of the vascular tone by EC will impair favoring vasoconstriction and further tissular hypoxia. The nutrient transfer to the fetus will alter on this condition, exposing it constantly to hyperglycemia. Persistent hyperglycemia will damage its blood vessels and force its β-cells to secrete insulin extensively, causing metabolic and vascular impairment that will predispose it to CVD before its birth.

Advertisement

Acknowledgments

Especial thanks to Tamara Jiménez Iturrieta for the illustrations.

The author declares that no funding was received for this publication.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Nomenclature

References

  1. 1. McAloon CJ, Boylan LM, Hamborg T, Stallard N, Osman F, Lim PB, et al. The changing face of cardiovascular disease 2000-2012: An analysis of the world health organisation global health estimates data. International Journal of Cardiology. 2016;224:256-264
  2. 2. National Heart, Lung and blood I. “What Are the Risk Factors for Heart Disease?” [Internet]. 2017. Available from: https://www.nhlbi.nih.gov/health-topics/education-and-awareness/heart-truth
  3. 3. Cosselman KE, Navas-acien A, Kaufman JD. Environmental factors in cardiovascular disease. Nature Reviews. Cardiology. 2015;12(November):627-642
  4. 4. Hakim J, Senterman MK, Hakim AM. Preeclampsia is a biomarker for vascular disease in both mother and child: The need for a medical alert system. Kumar P, editor. International Journal of Pediatrics. 2013;2013:953150. DOI: 10.1155/2013/953150
  5. 5. Napoli C, Glass CK, Witztum JL, Deutsch R, D’Armiento FP, Palinski W. Influence of maternal hypercholesterolaemia during pregnancy on progression of early atherosclerotic lesions in childhood: Fate of early lesions in children (FELIC) study. Lancet. 1999;354:1234-1241
  6. 6. Pathirana MM, Lassi ZS, Roberts CT, Andraweera PH. Cardiovascular risk factors in offspring exposed to gestational diabetes mellitus in utero: Systematic review and meta-analysis. Journal of Developmental Origins of Health and Disease. 2019;11(October):599-616
  7. 7. Benschop L, Duvekot JJ, Van Lennep JER. Future risk of cardiovascular disease risk factors and events in women after a hypertensive disorder of pregnancy. Heart (British Cardiac Society). 2019;76:1273-1278
  8. 8. Kramer CK, Campbell S, Retnakaran R. Gestational diabetes and the risk of cardiovascular disease in women: A systematic review and meta-analysis. Diabetologia. 2019;62(6):905-914
  9. 9. Napoli C, D’Armiento FP, Mancini FP, Postiglione A, Witztum JL, Palumbo G, et al. Fatty streak formation occurs in human fetal aortas and is greatly enhanced by maternal hypercholesterolemia. Intimal accumulation of low density lipoprotein and its oxidation precede monocyte recruitment into early atherosclerotic lesions. The Journal of Clinical Investigation. 1997;100(11):2680-2690
  10. 10. Cantin C, Fuenzalida B, Leiva A. Maternal hypercholesterolemia during pregnancy: Potential modulation of cholesterol transport through the human placenta and lipoprotein profile in maternal and neonatal circulation. Placenta. 2020;94(December):26-33
  11. 11. Leiva A, Salsoso R, Sáez T, Sanhueza C, Pardo F, Sobrevia L. Cross-sectional and longitudinal lipid determination studies in pregnant women reveal an association between increased maternal LDL cholesterol concentrations and reduced human umbilical vein relaxation. Placenta. 2015;36(8):895-902
  12. 12. Yang Y, Le Ray I, Zhu J, Zhang J, Hua J, Reilly M. Preeclampsia prevalence, risk factors, and pregnancy outcomes in Sweden and China. JAMA Network Open. 2021;4(5):e218401-e218401. DOI: 10.1001/jamanetworkopen.2021.8401
  13. 13. Mamun AA, Kinarivala MK, O’Callaghan M, Williams G, Najman J, Callaway L. Does hypertensive disorder of pregnancy predict offspring blood pressure at 21 years? Evidence from a birth cohort study. Journal of Human Hypertension [Internet]. 2012;26(5):288-294. DOI: 10.1038/jhh.2011.35
  14. 14. Davis EF, Lazdam M, Lewandowski AJ, Worton SA, Kelly B, Kenworthy Y, et al. Cardiovascular risk factors in children and young adults born to preeclamptic pregnancies: A systematic review. Pediatrics. 2012;129(6):e1552-e1561
  15. 15. Odegård RA, Vatten LJ, Nilsen ST, Salvesen KA, Austgulen R. Preeclampsia and fetal growth. Obstetrics and Gynecology. 2000;96(6):950-955
  16. 16. Menendez-Castro C, Rascher W, Hartner A. Intrauterine growth restriction - impact on cardiovascular diseases later in life. Molecular and Cellular Pediatrics. 2018;5(1):4. Available from: https://pubmed.ncbi.nlm.nih.gov/29560535
  17. 17. Zhu Y, Zhang C. Prevalence of gestational diabetes and risk of progression to type 2 Diabetes: A global perspective. Current Diabetes Reports. 2016;16(1):7. Available from: https://pubmed.ncbi.nlm.nih.gov/26742932
  18. 18. Lavery JA, Friedman AM, Keyes KM, Wright JD, Ananth CV. Gestational diabetes in the United States: Temporal changes in prevalence rates between 1979 and 2010. BJOG: An International Journal of Obstetrics & Gynaecology. 2017;124(5):804-813. DOI: 10.1111/1471-0528.14236
  19. 19. Leng J, Shao P, Zhang C, Tian H, Zhang F, Zhang S, et al. Prevalence of gestational diabetes mellitus and its risk factors in Chinese pregnant women: A prospective population-based study in Tianjin, China. PLoS One. 2015;10(3):e0121029
  20. 20. Ferrara A. Increasing prevalence of gestational diabetes mellitus. Diabetes Care [Internet]. 2007;30(Supplement 2):S141-S146. Available from: http://care.diabetesjournals.org/content/30/Supplement_2/S141.abstract
  21. 21. Collier A, Abraham EC, Armstrong J, Godwin J, Monteath K, Lindsay R. Reported prevalence of gestational diabetes in Scotland: The relationship with obesity, age, socioeconomic status, smoking and macrosomia, and how many are we missing? Journal of Diabetes Investigation. 2017;8(2):161-167
  22. 22. Yu Y, Arah OA, Liew Z, Cnattingius S, Olsen J, Sørensen HT, et al. Maternal diabetes during pregnancy and early onset of cardiovascular disease in offspring: Population based cohort study with 40 years of follow-up. BMJ. 2019;367:1-4
  23. 23. Lee J, Ouh Y-T, Ahn KH, Hong SC, Oh M-J, Kim H-J, et al. Preeclampsia: A risk factor for gestational diabetes mellitus in subsequent pregnancy. PLoS One. 2017;12(5):e0178150-e0178150 Available from: https://pubmed.ncbi.nlm.nih.gov/28542483
  24. 24. Plows JF, Stanley JL, Baker PN, Reynolds CM, Vickers MH. The pathophysiology of gestational diabetes mellitus. International Journal of Molecular Sciences. 2018;19(11):3342. Available from: https://pubmed.ncbi.nlm.nih.gov/30373146
  25. 25. Contreras-Duarte S, Carvajal L, Fuenzalida B, Cantin C, Sobrevia L, Leiva A. Maternal dyslipidaemia in pregnancy with gestational diabetes mellitus: Possible impact on Foetoplacental vascular function and lipoproteins in the neonatal circulation. Current Vascular Pharmacology. 2017;17(1):52-71
  26. 26. Espinoza C, Fuenzalida B, Leiva A. Increased fetal cardiovascular disease risk: Potential synergy between gestational diabetes mellitus and maternal hypercholesterolemia. Current Vascular Pharmacology. 2021;19:1-23
  27. 27. Classification and diagnosis of diabetes: Standards of medical care in diabetes-2018. Diabetes Care. 2018;41(Suppl. 1):S13-S27
  28. 28. Yang G-R, Dye TD, Li D. Effects of pre-gestational diabetes mellitus and gestational diabetes mellitus on macrosomia and birth defects in upstate New York. Diabetes Research and Clinical Practice. 2019;155:107811
  29. 29. Di Cianni G, Miccoli R, Volpe L, Lencioni C, Del Prato S. Intermediate metabolism in normal pregnancy and in gestational diabetes. Diabetes/Metabolism Research and Reviews. 2003;19(4):259-270
  30. 30. Catalano PM. Trying to understand gestational diabetes. Diabetic Medicine [Internet]. 2014;31(3):273-281 Available from: https://pubmed.ncbi.nlm.nih.gov/24341419
  31. 31. Catalano PM, Tyzbir ED, Roman NM, Amini SB, Sims EA. Longitudinal changes in insulin release and insulin resistance in nonobese pregnant women. American Journal of Obstetrics and Gynecology. 1991;165(6 Pt 1):1667-1672
  32. 32. Barbour LA, McCurdy CE, Hernandez TL, Kirwan JP, Catalano PM, Friedman JE. Cellular mechanisms for insulin resistance in normal pregnancy and gestational diabetes. Diabetes Care. 2007;30(Suppl. 2):S112-S119
  33. 33. Lain KY, Catalano PM. Metabolic changes in pregnancy. Clinical Obstetrics and Gynecology. 2007;50(4):938-948
  34. 34. Friedman JE, Ishizuka T, Shao J, Huston L, Highman T, Catalano P. Impaired glucose transport and insulin receptor tyrosine phosphorylation in skeletal muscle from obese women with gestational diabetes. Diabetes. 1999;48(9):1807-1814
  35. 35. Ryckman KK, Spracklen CN, Smith CJ, Robinson JG, Saftlas AF. Maternal lipid levels during pregnancy and gestational diabetes: A systematic review and meta-analysis. BJOG: An International Journal of Obstetrics and Gynaecology. 2015;122(5):643-651
  36. 36. Wang J, Li Z, Lin L. Maternal lipid profiles in women with and without gestational diabetes mellitus. Medicine (Baltimore). 2019;98(16):e15320-e15320. Available from: https://pubmed.ncbi.nlm.nih.gov/31008986
  37. 37. Leiva A, Pardo F, Ramírez MA, Farías M, Casanello P, Sobrevia L. Fetoplacental vascular endothelial dysfunction as an early phenomenon in the programming of human adult diseases in subjects born from gestational diabetes mellitus or obesity in pregnancy. Experimental Diabetes Research. 2011;2011:349286
  38. 38. Marco LJ, McCloskey K, Vuillermin PJ, Burgner D, Said J, Ponsonby AL. Cardiovascular disease risk in the offspring of diabetic women: The impact of the intrauterine environment. Experimental Diabetes Research. 2012;2012:565160
  39. 39. Huhn EA, Rossi SW, Hoesli I, Göbl CS. Controversies in screening and diagnostic criteria for gestational diabetes in early and late pregnancy. Frontiers in Endocrinology (Lausanne). 2018;9:696. DOI: https://www.frontiersin.org/article/10.3389/fendo.2018.00696
  40. 40. Buchanan TA, Xiang AH. Gestational diabetes mellitus. Journal of Clinical Investigation. 2005;115(3):485-491. Available from: https://pubmed.ncbi.nlm.nih.gov/15765129
  41. 41. Association AD. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2010;33(Supplement 1):S62-S69 Available from: https://care.diabetesjournals.org/content/33/Supplement_1/S62
  42. 42. Shang M, Lin L. IADPSG criteria for diagnosing gestational diabetes mellitus and predicting adverse pregnancy outcomes. Journal of Perinatology. 2014;34(2):100-104
  43. 43. Duran A, Sáenz S, Torrejón MJ, Bordiú E, del Valle L, Galindo M, et al. Introduction of IADPSG criteria for the screening and diagnosis of gestational diabetes mellitus results in improved pregnancy outcomes at a lower cost in a large Cohort of pregnant women: The St. Carlos gestational diabetes study. Diabetes Care. 2014;37(9):2442-2450. Available from: https://care.diabetesjournals.org/content/37/9/2442
  44. 44. Association AD. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes—2020. Diabetes Care. 2020;43(Supplement 1):S14-S31. Available from: https://care.diabetesjournals.org/content/43/Supplement_1/S14
  45. 45. Yang X, Hsu-Hage B, Zhang H, Zhang C, Zhang Y, Zhang C. Women with impaired glucose tolerance during pregnancy have significantly poor pregnancy outcomes. Diabetes Care. 2002;25(9):1619-1624
  46. 46. Araujo Júnior E, Peixoto AB, Zamarian ACP, Elito Júnior J, Tonni G. Macrosomia. Best Practice & Research. Clinical Obstetrics & Gynaecology. 2017;38:83-96
  47. 47. Fong A, Serra A, Herrero T, Pan D, Ogunyemi D. Pre-gestational versus gestational diabetes: A population based study on clinical and demographic differences. Journal of Diabetes and its Complications. 2014;28(1):29-34
  48. 48. Olmos PR, Rigotti A, Busso D, Berkowitz L, Santos JL, Borzone GR, et al. Maternal hypertriglyceridemia: A link between maternal overweight-obesity and macrosomia in gestational diabetes. Obesity (Silver Spring). 2014;22(10):2156-2163
  49. 49. Schaefer UM, Songster G, Xiang A, Berkowitz K, Buchanan TA, Kjos SL. Congenital malformations in offspring of women with hyperglycemia first detected during pregnancy. American Journal of Obstetrics and Gynecology. 1997;177(5):1165-1171
  50. 50. Ramírez-Emiliano J, Fajardo-Araujo ME, Zúñiga-Trujillo I, Pérez-Vázquez V, Sandoval-Salazar C, Órnelas-Vázquez JK. Mitochondrial content, oxidative, and nitrosative stress in human full-term placentas with gestational diabetes mellitus. Reproductive Biology and Endocrinology. 2017;15(1):26. DOI: 10.1186/s12958-017-0244-7
  51. 51. Lappas M, Hiden U, Desoye G, Froehlich J, Hauguel-de Mouzon S, Jawerbaum A. The role of oxidative stress in the pathophysiology of gestational diabetes mellitus. Antioxidants & Redox Signaling. 2011;15(12):3061-3100
  52. 52. Desoye G, Hauguel-De MS. The human placenta in gestational diabetes mellitus: The insulin and cytokine network. Diabetes Care. 2007;30(Suppl. 2):S120-S126
  53. 53. Zhu C, Yang H, Geng Q , Ma Q , Long Y, Zhou C, et al. Association of oxidative stress biomarkers with gestational diabetes mellitus in pregnant women: A case-control study. PLoS One. 2015;10(4):e0126490
  54. 54. Coughlan MT, Vervaart PP, Permezel M, Georgiou HM, Rice GE. Altered placental oxidative stress status in gestational diabetes mellitus. Placenta. 2004;25(1):78-84
  55. 55. Lingappan K. NF-κB in oxidative stress. Current Opinion in Toxicology. 2018;7:81-86
  56. 56. Radaelli T, Varastehpour A, Catalano P, Mouzon SH. Gestational diabetes induces placental genes for chronic stress and inflammatory pathways GDM alters expression profile of placental genes. Diabetes. 2003;52(September):2951-2958
  57. 57. Pessler D, Rudich A, Bashan N. Oxidative stress impairs nuclear proteins binding to the insulin responsive element in the GLUT4 promoter. Diabetologia. 2001;44(12):2156-2164
  58. 58. Turco MY, Moffett A. Development of the human placenta. Development. 2019;146:22
  59. 59. Knöfler M, Pollheimer J. Human placental trophoblast invasion and differentiation: A particular focus on Wnt signaling. Frontiers in Genetics. 2013;4:190. DOI: https://www.frontiersin.org/article/10.3389/fgene.2013.00190
  60. 60. Gude NM, Roberts CT, Kalionis B, King RG. Growth and function of the normal human placenta. Thrombosis Research. 2004;114(5-6 SPEC. ISS):397-407
  61. 61. Cross JC, Nakano H, Natale DRC, Simmons DG, Watson ED. Branching morphogenesis during development of placental villi. Differentiation. 2006;74(7):393-401
  62. 62. Pijnenborg R, Vercruysse L, Hanssens M. The uterine spiral arteries in human pregnancy: Facts and controversies. Placenta. 2006;27(9-10):939-958
  63. 63. Mayama R, Izawa T, Sakai K, Suciu N, Iwashita M. Improvement of insulin sensitivity promotes extravillous trophoblast cell migration stimulated by insulin-like growth factor-I. Endocrine Journal. 2013;60(3):359-368
  64. 64. Aires MB. Effects of maternal diabetes on trophoblast cells. World Journal of Diabetes. 2015;6(2):338
  65. 65. Edu A, Teodorescu C, Dobjanschi CG, Socol ZZ, Teodorescu V, Matei A, et al. Placenta changes in pregnancy with gestational diabetes. Rom J Morphol Embryol = Revue Roumaine de Morphologie et d'Embryologie. 2016;57(2):507-512
  66. 66. Taricco E, Radaelli T, Nobile de Santis MS, Cetin I. Foetal and placental weights in relation to maternal characteristics in gestational diabetes. Placenta. 2003;24(4):343-347
  67. 67. Huynh J, Dawson D, Roberts D, Bentley-Lewis R. A systematic review of placental pathology in maternal diabetes mellitus. Placenta. 2015;36(2):101-114
  68. 68. Loegl J, Nussbaumer E, Cvitic S, Huppertz B, Desoye G, Hiden U. GDM alters paracrine regulation of feto-placental angiogenesis via the trophoblast. Laboratory Investigation. 2017;97(4):409-418
  69. 69. Unek G, Ozmen A, Mendilcioglu I, Simsek M, Korgun ET. Immunohistochemical distribution of cell cycle proteins p27, p57, cyclin D3, PCNA and Ki67 in normal and diabetic human placentas. Journal of Molecular Histology. 2014;45(1):21-34
  70. 70. Bandres-Meriz J, Hoch D, Honeder S, Majali-Martinez A, Emilio H, Matthias S, et al. 93-OR: Low maternal Insulin sensitivity associates with DNA-related functions in human first-trimester trophoblast. Diabetes. 2020;69(Supplement 1). Available from: https://diabetes.diabetesjournals.org/content/69/Supplement_1/93-OR
  71. 71. Vega M, Mauro M, Williams Z. Direct toxicity of insulin on the human placenta and protection by metformin. Fertility and Sterility. 2019;111(3):489-496.e5
  72. 72. Smith SC, Leung TN, To KF, Baker PN. Apoptosis is a rare event in first-trimester placental tissue. American Journal of Obstetrics and Gynecology. 2000;183(3):697-699
  73. 73. Sharp AN, Heazell AEP, Crocker IP, Mor G. Placental apoptosis in health and disease. American Journal of Reproductive Immunology. 2010;64(3):159-169. Available from: https://pubmed.ncbi.nlm.nih.gov/20367628
  74. 74. Magee TR, Ross MG, Wedekind L, Desai M, Kjos S, Belkacemi L. Gestational diabetes mellitus alters apoptotic and inflammatory gene expression of trophobasts from human term placenta. Journal of Diabetes and its Complications. 2014;28(4):448-459
  75. 75. Sgarbosa F, Barbisan LF, Brasil MAM, Costa E, Calderon IMP, Gonçalves CR, et al. Changes in apoptosis and Bcl-2 expression in human hyperglycemic, term placental trophoblast. Diabetes Research and Clinical Practice. 2006;73(2):143-149
  76. 76. Westermeier F, Sáez T, Arroyo P, Toledo F, Gutiérrez J, Sanhueza C, et al. Insulin receptor isoforms: An integrated view focused on gestational diabetes mellitus. Diabetes/Metabolism Research and Reviews. 2016;32(4):350-365
  77. 77. Benirschke K, Kaufmann PB, R. Pathology of the Humanplacenta. 5th ed. New York: Springer; 2006
  78. 78. Villalobos-Labra R, Silva L, Subiabre M, Araos J, Salsoso R, Fuenzalida B, et al. Akt/mTOR role in human Foetoplacental vascular Insulin resistance in diseases of pregnancy. Wadsack C, editor. Journal of Diabetes Research. 2017;2017:5947859. DOI: 10.1155/2017/5947859
  79. 79. Colomiere M, Permezel M, Riley C, Desoye G, Lappas M. Defective insulin signaling in placenta from pregnancies complicated by gestational diabetes mellitus. European Journal of Endocrinology. 2009;160(4):567-578
  80. 80. Pérez-Pérez A, Guadix P, Maymó J, Dueñas JL, Varone C, Fernández-Sánchez M, et al. Insulin and leptin signaling in placenta from gestational diabetic subjects. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et métabolisme. 2016;48(1):62-69
  81. 81. Subiabre M, Silva L, Toledo F, Paublo M, López MA, Boric MP, et al. Insulin therapy and its consequences for the mother, foetus, and newborn in gestational diabetes mellitus. Biochimica et Biophysica Acta (BBA)—Molecular Basis of Disease. 2018;1864(9):2949-2956. DOI: 10.1016/j.bbadis.2018.06.005
  82. 82. Na K-H, Choi JH, Kim C-H, Kim K-S, Kim GJ. Altered expression of norepinephrine transporter and norepinephrine in human placenta cause pre-eclampsia through regulated trophoblast invasion. Clinical and Experimental Reproductive Medicine. 2013;40(1):12-22. Available from: https://pubmed.ncbi.nlm.nih.gov/23614111
  83. 83. Opichka MA, Rappelt MW, Gutterman DD, Grobe JL, McIntosh JJ. Review vascular dysfunction in preeclampsia. Cells. 2021;10:11
  84. 84. Savvidou MD, Syngelaki A, Balakitsas N, Panaiotova E, Nicolaides KH. First-trimester uterine artery Doppler examination in pregnancies complicated by gestational diabetes mellitus with or without pre-eclampsia. Ultrasound in Obstetrics & Gynecology (UOG), the official journal of the International Society of Ultrasound in Obstetrics and Gynecology. 2013;42(5):525-529
  85. 85. Istrate-Ofiţeru AM, Berceanu C, Berceanu S, Busuioc CJ, Roşu GC, Diţescu D, et al. The influence of gestational diabetes mellitus (GDM) and gestational hypertension (GH) on placental morphological changes. Romanian Journal of Morphology and Embryology. 2020;61(2):371-384 Available from: https://pubmed.ncbi.nlm.nih.gov/33544789
  86. 86. Downing JW, Baysinger CL, Johnson RF, Paschall RL, Shotwell MS. The effects of vasopressin and oxytocin on the fetoplacental distal stem arteriolar vascular resistance of the dual-perfused, single, isolated, human placental cotyledon. Anesthesia & Analgesia. 2016;123(3):698-702
  87. 87. Stevens AD, Lumbers ER. Effects of intravenous infusions of noradrenaline into the pregnant ewe on uterine blood flow, fetal renal function, and lung liquid flow. Canadian Journal of Physiology and Pharmacology. 1995;73(2):202-208
  88. 88. Christensen NJ. Plasma norepinephrine and epinephrine in untreated diabetics, during fasting and after insulin administration. Diabetes. 1974;23(1):1-8. DOI: 10.2337/diab.23.1.1
  89. 89. Feng Y, Feng Q , Qu H, Song X, Hu J, Xu X, et al. Stress adaptation is associated with insulin resistance in women with gestational diabetes mellitus. Nutrition & Diabetes. 2020;10(1):4
  90. 90. Levin BE. Glucose increases rat plasma norepinephrine levels by direct action on the brain. The American Journal of Physiology. 1991;261(6 Pt 2):R1351-R1357
  91. 91. Walters JM, Ward GM, Barton J, Arackal R, Boston RC, Best JD, et al. The effect of norepinephrine on insulin secretion and glucose effectiveness in non-insulin-dependent diabetes. Metabolism. 1997;46(12):1448-1453
  92. 92. Baron AD, Brechtel G, Johnson A, Fineberg N, Henry DP, Steinberg HO. Interactions between insulin and norepinephrine on blood pressure and insulin sensitivity. Studies in lean and obese men. Journal of Clinical Investigation. 1994;93(6):2453-2462. Available from: https://pubmed.ncbi.nlm.nih.gov/8200981
  93. 93. Hiatt N, Davidson MB, Chapman LW, Sheinkopf JA. Epinephrine enhancement of potassium-stimulated immunoreactive insulin secretion. Role of beta-adrenergic receptors. Diabetes. 1978;27(5):550-553
  94. 94. Yart L, Roset Bahmanyar E, Cohen M, Martinez de Tejada B. Role of the uteroplacental renin-angiotensin system in placental development and function, and its implication in the preeclampsia pathogenesis. Biomedicines. 2021;9(10):1332. Available from: https://pubmed.ncbi.nlm.nih.gov/34680449
  95. 95. Kalenga MK, Thomas K, De Gasparo M, De Hertogh R. Determination of renin, angiotensin converting enzyme and angiotensin II levels in human placenta, chorion and amnion from women with pregnancy induced hypertension. Clinical Endocrinology. 1996;44(4):429-433. DOI: 10.1046/j.1365-2265.1996.703525.x
  96. 96. Delforce SJ, Wang Y, Van-Aalst ME, Corbisier de Meaultsart C, Morris BJ, Broughton-Pipkin F, et al. Effect of oxygen on the expression of renin-angiotensin system components in a human trophoblast cell line. Placenta. 2016;37:1-6
  97. 97. Cooper AC, Robinson G, Vinson GP, Cheung WT, Broughton PF. The localization and expression of the renin-angiotensin system in the human placenta throughout pregnancy. Placenta. 1999;20(5-6):467-474
  98. 98. Kurlak LO, Mistry HD, Cindrova-Davies T, Burton GJ, Broughton PF. Human placental renin-angiotensin system in normotensive and pre-eclamptic pregnancies at high altitude and after acute hypoxia-reoxygenation insult. The Journal of Physiology. 2016;594(5):1327-1340
  99. 99. Velazquez-Roman JA, Villafaña S, Lopez Sanchez P, Fernandez-Vallín E, Bobadilla Lugo RA. Effect of pregnancy and diabetes on vascular receptors for angiotensin II. Clinical and Experimental Hypertension. 2011;33(3):167-173
  100. 100. Stevens AD, Lumbers ER. The effects of long-term infusions of angiotensin II into the pregnant ewe on uterine blood flow and on the fetus. Journal of Cardiovascular Pharmacology. 1999;34(6):824-830
  101. 101. Samuelsson A-M, Bollano E, Mobini R, Larsson B-M, Omerovic E, Fu M, et al. Hyperinsulinemia: Effect on cardiac mass/function, angiotensin II receptor expression, and insulin signaling pathways. American Journal of Physiology—Heart and Circulatory Physiology. 2006;291(2):H787-H796. DOI: 10.1152/ajpheart.00974.2005
  102. 102. Tuck ML, Bounoua F, Eslami P, Nyby MD, Eggena P, Corry DB. Insulin stimulates endogenous angiotensin II production via a mitogen-activated protein kinase pathway in vascular smooth muscle cells. Journal of Hypertension. 2004;22(9):1779-1785. Available from: https://journals.lww.com/jhypertension/Fulltext/2004/09000/Insulin_stimulates_endogenous_angiotensin_II.23.aspx
  103. 103. Watanabe N, Morimoto S, Fujiwara T, Suzuki T, Taniguchi K, Mori F, et al. Prediction of gestational diabetes mellitus by soluble (pro)renin receptor during the first trimester. The Journal of Clinical Endocrinology and Metabolism. 2013;98(6):2528-2535
  104. 104. Ribeiro-Oliveira A Jr, Nogueira AI, Pereira RM, Boas WWV, Dos Santos RAS, Simões e Silva AC. The renin-angiotensin system and diabetes: An update. Vascular Health and Risk Management. 2008;4(4):787-803. Available from: https://pubmed.ncbi.nlm.nih.gov/19065996
  105. 105. Chu KY, Leung PS. Angiotensin II in type 2 diabetes mellitus. Current Protein & Peptide Science. 2009;10(1):75-84
  106. 106. Tufiño C, Villanueva-López C, Ibarra-Barajas M, Bracho-Valdés I, Bobadilla-Lugo RA. Experimental gestational diabetes mellitus induces blunted vasoconstriction and functional changes in the rat aorta. BioMed Research International. 2014;2014:329634
  107. 107. Chen Y-P, Li J, Wang Z-N, Reichetzeder C, Xu H, Gong J, et al. Renin angiotensin aldosterone system and glycemia in pregnancy. Clinical Laboratory. 2012;58(5-6):527-533
  108. 108. Chen Z-W, Tsai C-H, Pan C-T, Chou C-H, Liao C-W, Hung C-S, et al. Endothelial dysfunction in primary Aldosteronism. International Journal of Molecular Sciences. 2019;20(20):5214. Available from: https://pubmed.ncbi.nlm.nih.gov/31640178
  109. 109. Zhang F, Xiao X, Liu D, Dong X, Sun J, Zhang X. Increased cord blood angiotensin II concentration is associated with decreased insulin sensitivity in the offspring of mothers with gestational diabetes mellitus. Journal of Perinatology. 2013;33(1):9-14. DOI: 10.1038/jp.2012.40
  110. 110. Wei Y, Sowers JR, Nistala R, Gong H, Uptergrove GME, Clark SE, et al. Angiotensin II-induced NADPH oxidase activation impairs insulin signaling in skeletal muscle cells. Journal of Biological Chemistry. 2006;281(46):35137-35146. DOI: 10.1074/jbc.M601320200
  111. 111. Olivares-Reyes JA, Arellano-Plancarte A, Castillo-Hernandez JR. Angiotensin II and the development of insulin resistance: Implications for diabetes. Molecular and Cellular Endocrinology. 2009;302(2):128-139
  112. 112. Aroor AR, DeMarco VG, Jia G, Sun Z, Nistala R, Meininger GA, et al. The role of tissue renin-angiotensin-aldosterone system in the development of endothelial dysfunction and arterial stiffness. Frontiers in Endocrinology (Lausanne). 2013;4(October):1-7. DOI: http://journal.frontiersin.org/article/10.3389/fendo.2013.00161/abstract
  113. 113. Hurrle S, Hsu WH. The etiology of oxidative stress in insulin resistance. Biomedical Journal. 2017;40(5):257-262. Available from: https://pubmed.ncbi.nlm.nih.gov/29179880
  114. 114. Irani RA, Zhang Y, Blackwell SC, Zhou CC, Ramin SM, Kellems RE, et al. The detrimental role of angiotensin receptor agonistic autoantibodies in intrauterine growth restriction seen in preeclampsia. The Journal of Experimental Medicine. 2009;206(12):2809-2822
  115. 115. Widmer RJ, Lerman A. Endothelial dysfunction and cardiovascular disease. Gobal Cardiology Science & Practice. 2014;2014(3):291-308. Available from: https://pubmed.ncbi.nlm.nih.gov/25780786
  116. 116. Lerman A, Burnett JCJ. Intact and altered endothelium in regulation of vasomotion. Circulation. 1992;86(Suppl. 6):III12-III19
  117. 117. Voynow JA, Kummarapurugu A. Isoprostanes and asthma. Biochimica et Biophysica Acta. 2011;1810(11):1091-1095
  118. 118. Kuhn DC, Botti JJ, Cherouny PH, Demers LM. Eicosanoid production and transfer in the placenta of the diabetic pregnancy. Prostaglandins. 1990;40(2):205-215
  119. 119. Wang Y, Walsh SW, Kay HH. Placental lipid peroxides and thromboxane are increased and prostacyclin is decreased in women with preeclampsia. American Journal of Obstetrics and Gynecology. 1992;167(4 Pt 1):946-949
  120. 120. Ozkor MA, Quyyumi AA. Endothelium-derived hyperpolarizing factor and vascular function. Cardiology Research and Practice. 2011;2011:156146. Available from: https://pubmed.ncbi.nlm.nih.gov/21876822
  121. 121. Luksha L, Luksha N, Kublickas M, Nisell H, Kublickiene K. Diverse mechanisms of endothelium-derived hyperpolarizing factor-mediated dilatation in small myometrial arteries in normal human pregnancy and preeclampsia. Biology of Reproduction. 2010;83(5):728-735
  122. 122. Mokhtar SS, Vanhoutte PM, Leung SWS, Yusof MI, Wan Sulaiman WA, Mat Saad AZ, et al. Endothelium dependent hyperpolarization-type relaxation compensates for attenuated nitric oxide-mediated responses in subcutaneous arteries of diabetic patients. Nitric Oxide: Biology and Chemistry. 2016;53:35-44
  123. 123. San Martín R, Sobrevia L. Gestational diabetes and the adenosine/L-arginine/nitric oxide (ALANO) pathway in human umbilical vein endothelium. Placenta. 2006;27(1):1-10
  124. 124. Rafikov R, Fonseca FV, Kumar S, Pardo D, Darragh C, Elms S, et al. eNOS activation and NO function: Structural motifs responsible for the posttranslational control of endothelial nitric oxide synthase activity. The Journal of Endocrinology. 2011;210(3):271-284
  125. 125. Huang X, Liu G, Guo J, Su Z. The PI3K/AKT pathway in obesity and type 2 diabetes. International Journal of Biological Sciences. 2018;14(11):1483-1496. Available from: https://pubmed.ncbi.nlm.nih.gov/30263000
  126. 126. Chae I-H, Park K-W, Kim H-S, Oh B-H. Nitric oxide-induced apoptosis is mediated by Bax/Bcl-2 gene expression, transition of cytochrome c, and activation of caspase-3 in rat vascular smooth muscle cells. Clinica Chimica Acta. 2004;341(1-2):83-91
  127. 127. Tsihlis ND, Oustwani CS, Vavra AK, Jiang Q , Keefer LK, Kibbe MR. Nitric oxide inhibits vascular smooth muscle cell proliferation and neointimal hyperplasia by increasing the ubiquitination and degradation of UbcH10. Cell Biochemistry and Biophysics. 2011;60(1-2):89-97
  128. 128. Yaghini FA, Song CY, Lavrentyev EN, Ghafoor HUB, Fang XR, Estes AM, et al. Angiotensin II-induced vascular smooth muscle cell migration and growth are mediated by cytochrome P450 1B1-dependent superoxide generation. Hypertens. 2010;55(6):1461-1467. Available from: https://pubmed.ncbi.nlm.nih.gov/20439821
  129. 129. Tumminia A, Scalisi NM, Milluzzo A, Ettore G, Vigneri R, Sciacca L. Maternal diabetes impairs insulin and IGF-1 receptor expression and Signaling in human placenta. Frontiers in Endocrinology (Lausanne). 2021;12:119. Available from:. DOI: https://www.frontiersin.org/article/10.3389/fendo.2021.621680
  130. 130. Subiabre M, Silva L, Villalobos-Labra R, Toledo F, Paublo M, López MA, et al. Maternal insulin therapy does not restore foetoplacental endothelial dysfunction in gestational diabetes mellitus. Biochimica et Biophysica Acta—Molecular Basis of Disease. 2017;1863(11):2987-2998
  131. 131. Leiva A, Fuenzalida B, Barros E, Sobrevia B, Salsoso R, Sáez T, et al. Nitric oxide is a central common metabolite in vascular dysfunction associated with diseases of human pregnancy. Current Vascular Pharmacology. 2016;14(3):237-259
  132. 132. Grillo MA, Lanza A, Colombatto S. Transport of amino acids through the placenta and their role. Amino Acids. 2008;34(4):517-523
  133. 133. Subiabre M, Villalobos-Labra R, Silva L, Fuentes G, Toledo F, Sobrevia L. Role of insulin, adenosine, and adipokine receptors in the foetoplacental vascular dysfunction in gestational diabetes mellitus. Biochim Biophys Acta—Molecular Basis of Disease. 2020;1866(2):165370. DOI: 10.1016/j.bbadis.2018.12.021
  134. 134. Ballard-Croft C, Locklar AC, Keith BJ, Mentzer RMJ, Lasley RD. Oxidative stress and adenosine A1 receptor activation differentially modulate subcellular cardiomyocyte MAPKs. American Journal of Physiology. Heart and Circulatory Physiology. 2008;294(1):H263-H271
  135. 135. Guzmán-Gutiérrez E, Westermeier F, Salomón C, González M, Pardo F, Leiva A, et al. Insulin-increased L-arginine transport requires A(2A) adenosine receptors activation in human umbilical vein endothelium. PLoS One. 2012;7(7):e41705
  136. 136. Farías M, Puebla C, Westermeier F, Jo MJ, Pastor-Anglada M, Casanello P, et al. Nitric oxide reduces SLC29A1 promoter activity and adenosine transport involving transcription factor complex hCHOP-C/EBPalpha in human umbilical vein endothelial cells from gestational diabetes. Cardiovascular Research. 2010;86(1):45-54
  137. 137. Westermeier F, Salomón C, González M, Puebla C, Guzmán-Gutiérrez E, Cifuentes F, et al. Insulin restores gestational diabetes mellitus-reduced adenosine transport involving differential expression of insulin receptor isoforms in human umbilical vein endothelium. Diabetes. 2011;60(6):1677-1687
  138. 138. Feoktistov I, Goldstein AE, Ryzhov S, Zeng D, Belardinelli L, Voyno-Yasenetskaya T, et al. Differential expression of adenosine receptors in human endothelial cells. Circulation Research. 2002;90(5):531-538. DOI: 10.1161/01.RES.0000012203.21416.14
  139. 139. Razak AA, Leach L, Ralevic V. Impaired vasocontractile responses to adenosine in chorionic vessels of human term placenta from pregnant women with pre-existing and gestational diabetes. Diabetes & Vascular Disease Research. 2018;15(6):528-540
  140. 140. Baldea I, Teacoe I, Olteanu DE, Vaida-Voievod C, Clichici A, Sirbu A, et al. Effects of different hypoxia degrees on endothelial cell cultures-time course study. Mechanisms of Ageing and Development. 2018;172:45-50
  141. 141. Volpe CMO, Villar-Delfino PH, dos Anjos PMF, Nogueira-Machado JA. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death and Disease. 2018;9(2):119. DOI: 10.1038/s41419-017-0135-z
  142. 142. Sada K, Nishikawa T, Kukidome D, Yoshinaga T, Kajihara N, Sonoda K, et al. Hyperglycemia induces cellular hypoxia through production of mitochondrial ROS followed by suppression of aquaporin-1. PLoS One. 2016;11(7):e0158619. Available from: DOI: 10.1371/journal.pone.0158619
  143. 143. Sobrevia L, Valero P, Grismaldo A, Villalobos-Labra R, Pardo F, Subiabre M, et al. Mitochondrial dysfunction in the fetoplacental unit in gestational diabetes mellitus. Biochimica et Biophysica Acta: Molecular Basis of Disease. 2020;1866(12):165948. DOI: 10.1016/j.bbadis.2020.165948
  144. 144. Fisher JJ, Vanderpeet CL, Bartho LA, McKeating DR, Cuffe JSM, Holland OJ, et al. Mitochondrial dysfunction in placental trophoblast cells experiencing gestational diabetes mellitus. The Journal of Physiology. 2021;599(4):1291-1305
  145. 145. Lappas M, Mitton A, Permezel M. In response to oxidative stress, the expression of inflammatory cytokines and antioxidant enzymes are impaired in placenta, but not adipose tissue, of women with gestational diabetes. The Journal of Endocrinology. 2010;204(1):75-84
  146. 146. Brown GC. Nitric oxide and mitochondrial respiration. Biochimica et Biophysica Acta. 1999;1411(2-3):351-369
  147. 147. Eppel D, Feichtinger M, Lindner T, Kotzaeridi G, Rosicky I, Yerlikaya-Schatten G, et al. Association between maternal triglycerides and disturbed glucose metabolism in pregnancy. Acta Diabetologica. 2021;58(4):459-465
  148. 148. Dubé M-C, Morrisset A-S, Tchernof A, Weisnagel SJ. Cord blood C-peptide levels relate to the metabolic profile of women with and without gestational diabetes. Acta Obstetricia et Gynecologica Scandinavica. 2012;91(12):1469-1473. DOI: 10.1111/aogs.12005
  149. 149. Eslamian L, Akbari S, Marsoosi V, Jamal A. Association between fetal overgrowth and metabolic parameters in cord blood of newborns of women with GDM. Minerva Medica. 2013;104(3):317-324
  150. 150. Son GH, Kwon JY, Kim YH, Park YW. Maternal serum triglycerides as predictive factors for large-for-gestational age newborns in women with gestational diabetes mellitus. Acta Obstetricia et Gynecologica Scandinavica. 2010;89(5):700-704
  151. 151. Herrera E, Ortega-Senovilla H. Disturbances in lipid metabolism in diabetic pregnancy—Are these the cause of the problem? Best Practice & Research Clinical Endocrinology & Metabolism. 2010;24(4):515-525. DOI: 10.1016/j.beem.2010.05.006
  152. 152. Abbassi-Ghanavati M, Greer LG, Cunningham FG. Pregnancy and laboratory studies: A reference table for clinicians. Obstetrics and Gynecology. 2009;114(6):1326-1331
  153. 153. Villafan-Bernal JR, Acevedo-Alba M, Reyes-Pavon R, Diaz-Parra GA, Lip-Sosa DL, Vazquez-Delfin HI, et al. Plasma levels of free fatty acids in women with gestational diabetes and its intrinsic and extrinsic determinants: Systematic review and meta-analysis. Journal of Diabetes Research. 2019;2019:7098470. Available from: https://pubmed.ncbi.nlm.nih.gov/31531374
  154. 154. Bomba-Opon D, Wielgos M, Szymanska M, Bablok L. Effects of free fatty acids on the course of gestational diabetes mellitus. Neuro Endocrinology Letters. 2006;27(1-2):277-280
  155. 155. Kajikawa M, Higashi Y. Triglycerides and endothelial function: Molecular biology to clinical perspective. Current Opinion in Lipidology. 2019;30(5):364-369. Available from: https://journals.lww.com/co-lipidology/Fulltext/2019/10000/Triglycerides_and_endothelial_function__molecular.3.aspx
  156. 156. Milosavljevic D, Kontush A, Griglio S, Le Naour G, Thillet J, Chapman MJ. VLDL-induced triglyceride accumulation in human macrophages is mediated by modulation of LPL lipolytic activity in the absence of change in LPL mass. Biochimica et Biophysica Acta. 2003;1631(1):51-60
  157. 157. Wang L, Gill R, Pedersen TL, Higgins LJ, Newman JW, Rutledge JC. Triglyceride-rich lipoprotein lipolysis releases neutral and oxidized FFAs that induce endothelial cell inflammation. Journal of Lipid Research. 2009;50(2):204-213
  158. 158. Castillo-Castrejon M, Powell TL. Placental nutrient transport in gestational diabetic pregnancies. Frontiers in Endocrinology (Lausanne). 2017;8:306. Available from:. DOI: https://www.frontiersin.org/article/10.3389/fendo.2017.00306
  159. 159. Ericsson A, Hamark B, Powell TL, Jansson T. Glucose transporter isoform 4 is expressed in the syncytiotrophoblast of first trimester human placenta. Human Reproduction. 2005;20(2):521-530
  160. 160. Illsley NP. Glucose transporters in the human placenta. Placenta. 2000;21(1):14-22
  161. 161. Borges MH, Pullockaran J, Catalano P, Baumann MU, Illsley NP. Human placental GLUT1 glucose transporter expression and the fetal insulin-like growth factor axis in pregnancies complicated by diabetes. Biochimica et Biophysica Acta. Molecular Basis of Disease. 2019;1865(9):2411-2419
  162. 162. James-Allan LB, Arbet J, Teal SB, Powell TL, Jansson T. Insulin stimulates GLUT4 trafficking to the Syncytiotrophoblast basal plasma membrane in the human placenta. The Journal of Clinical Endocrinology and Metabolism. 2019;104(9):4225-4238
  163. 163. Díaz P, Dimasuay KG, Koele-Schmidt L, Jang B, Barbour LA, Jansson T, et al. Glyburide treatment in gestational diabetes is associated with increased placental glucose transporter 1 expression and higher birth weight. Placenta. 2017;57:52-59
  164. 164. Stanirowski PJ, Szukiewicz D, Pyzlak M, Abdalla N, Sawicki W, Cendrowski K. Impact of pre-gestational and gestational diabetes mellitus on the expression of glucose transporters GLUT-1, GLUT-4 and GLUT-9 in human term placenta. Endocrine [Internet]. 2017;55(3):799-808. Available from: https://pubmed.ncbi.nlm.nih.gov/27981520
  165. 165. Gaither K, Quraishi AN, Illsley NP. Diabetes alters the expression and activity of the human placental GLUT1 glucose transporter. The Journal of Clinical Endocrinology and Metabolism. 1999;84(2):695-701
  166. 166. Illsley NP, Sellers MC, Wright RL. Glycaemic regulation of glucose transporter expression and activity in the human placenta. Placenta. 1998;19(7):517-524
  167. 167. Hahn T, Barth S, Graf R, Engelmann M, Beslagic D, Reul JM, et al. Placental glucose transporter expression is regulated by glucocorticoids. The Journal of Clinical Endocrinology and Metabolism. 1999;84(4):1445-1452
  168. 168. Challier JC, Hauguel S, Desmaizieres V. Effect of insulin on glucose uptake and metabolism in the human placenta. The Journal of Clinical Endocrinology and Metabolism. 1986;62(5):803-807
  169. 169. Kamana KCS, Shakya ZH. Gestational diabetes mellitus and macrosomia: A literature review. Annals of Nutrition & Metabolism. 2015;66:14-20

Written By

Cristian Espinoza

Submitted: 03 January 2022 Reviewed: 12 January 2022 Published: 17 February 2022