Hostname: page-component-745bb68f8f-b6zl4 Total loading time: 0 Render date: 2025-02-06T10:38:49.654Z Has data issue: false hasContentIssue false

Endothelial dysfunction in individuals born after fetal growth restriction: cardiovascular and renal consequences and preventive approaches

Published online by Cambridge University Press:  02 May 2017

C. Yzydorczyk*
Affiliation:
Department Woman-Mother-Child, Clinic of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
J. B. Armengaud
Affiliation:
Department Woman-Mother-Child, Clinic of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
A. C. Peyter
Affiliation:
Department Woman-Mother-Child, Clinic of Neonatology, Neonatal Research Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
H. Chehade
Affiliation:
Department Woman-Mother-Child, Clinic of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland Department Woman-Mother-Child, Clinic of Pediatrics, Division of Pediatric Nephrology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
F. Cachat
Affiliation:
Department Woman-Mother-Child, Clinic of Pediatrics, Division of Pediatric Nephrology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
C. Juvet
Affiliation:
Department Woman-Mother-Child, Clinic of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland Department Woman-Mother-Child, Clinic of Pediatrics, Division of Pediatric Nephrology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
B. Siddeek
Affiliation:
Department Woman-Mother-Child, Clinic of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
S. Simoncini
Affiliation:
VRCM, Aix Marseille University, UMR S INSERM 1076, Faculté de Pharmacie, Marseille, France
F. Sabatier
Affiliation:
VRCM, Aix Marseille University, UMR S INSERM 1076, Faculté de Pharmacie, Marseille, France
F. Dignat-George
Affiliation:
VRCM, Aix Marseille University, UMR S INSERM 1076, Faculté de Pharmacie, Marseille, France
D. Mitanchez
Affiliation:
Division of Neonatology, Department of Perinatology, Armand Trousseau Hospital, APHP, Paris, France Sorbonne University, UPMC University Paris 06, Paris, France
U. Simeoni
Affiliation:
Department Woman-Mother-Child, Clinic of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
*
*Address for correspondence: C. Yzydorczyk, Department Woman-Mother-Child, Clinic of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland.(Email catherine.yzydorczyk@chuv.ch)
Rights & Permissions [Opens in a new window]

Abstract

Individuals born after intrauterine growth restriction (IUGR) have an increased risk of perinatal morbidity/mortality, and those who survive face long-term consequences such as cardiovascular-related diseases, including systemic hypertension, atherosclerosis, coronary heart disease and chronic kidney disease. In addition to the demonstrated long-term effects of decreased nephron endowment and hyperactivity of the hypothalamic–pituitary–adrenal axis, individuals born after IUGR also exhibit early alterations in vascular structure and function, which have been identified as key factors of the development of cardiovascular-related diseases. The endothelium plays a major role in maintaining vascular function and homeostasis. Therefore, it is not surprising that impaired endothelial function can lead to the long-term development of vascular-related diseases. Endothelial dysfunction, particularly impaired endothelium-dependent vasodilation and vascular remodeling, involves decreased nitric oxide (NO) bioavailability, impaired endothelial NO synthase functionality, increased oxidative stress, endothelial progenitor cells dysfunction and accelerated vascular senescence. Preventive approaches such as breastfeeding, supplementation with folate, vitamins, antioxidants, L-citrulline, L-arginine and treatment with NO modulators represent promising strategies for improving endothelial function, mitigating long-term outcomes and possibly preventing IUGR of vascular origin. Moreover, the identification of early biomarkers of endothelial dysfunction, especially epigenetic biomarkers, could allow early screening and follow-up of individuals at risk of developing cardiovascular and renal diseases, thus contributing to the development of preventive and therapeutic strategies to avert the long-term effects of endothelial dysfunction in infants born after IUGR.

Type
Review
Copyright
© Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2017 

We performed an extensive and critical review of the literature in order to explore the manifestations of endothelial dysfunction in individuals born after intrauterine growth restriction (IUGR) and examined which mechanisms may be incriminated and which preventive strategies could represent promising approaches. We used the following terms in the Pubmed library (MESH terms and free text), without time or language limits: (Prenatal Exposure Delayed Effects OR Late Effect, Prenatal Exposure OR Nutrition Disorders/Physiopathology OR Fetal Growth restriction) AND (Cardiovascular Diseases/Etiology OR Hypertension/Etiology) AND (Impaired Endothelial Function OR Oxidative Stress/Senescence). We included the most significant human and animal studies. From the references of the retrieved papers, additional articles were selected for this review. One author (C.Y.) read the titles and abstracts and selected the articles to be included.

IUGR: definition and risk factors

Definition

IUGR is defined as the inability of the fetus to reach its genetically determined potential size.Reference Brodsky and Christou 1 , Reference Jang, Jo, Lee, Kim and Lee 2 IUGR affects ~5–15% of all pregnancies in the United States and Europe, but its incidence varies widely and appears to be higher in low-income countries (it affects 30–55% of infants born in South Central Asia, 15–25% in Africa and 10–20% in Latin America).Reference Saleem, Sajjad and Fatima 3 Using the ReCoDe classification system, IUGR has been considered the most commonly identified factor in stillborn infants.Reference Mongelli and Gardosi 4 Therefore, the management of growth-restricted fetuses in terms of choosing the optimal delivery time is important to decrease perinatal mortality/morbidity. Fetal growth restriction is difficult to detect because of the lack of international consensus on the definition and diagnostic criteria for IUGR. In clinical practice, growth-restricted fetuses are usually identified based on birth weight (<10th percentile). However, some propose that using <3rd or <5th percentile as the criterion would better identify individuals at a higher risk of adverse perinatal outcomes. 5 Moreover, low estimated fetal weight (<10th percentile), certain ultrasound findings of fetal growth (abdominal circumference <2.5th percentile) and altered Doppler velocimetry indices, such as abnormal umbilical artery waveforms or decreased pulsatility of the middle cerebral artery, that suggest abnormalities in fetal circulation are also indicative of IUGR.Reference Alberry and Soothill 6 To better understand abnormal fetal growth and to detect IUGR, specific computerized fetal growth charts that consider fetal gender and maternal characteristics such as height, weight, parity and ethnic origin were developed by Gardosi et al. Reference Figueras and Gardosi 7 Pathological factors, including maternal systemic hypertension (HTN), diabetes, tobacco use and preterm delivery, were excluded from the model to predict the optimum weight that a baby can reach at term during a normal pregnancy. As it is necessary to distinguish between growth-restricted and constitutive ‘small for gestational age’ fetuses, longitudinal assessments of fetal growth trajectories are required to identify pathological fetal growth restriction, even if the altered growth trajectory is above the 10th centile limit.Reference Barker, McAuliffe and Alderdice 8 More recently, universal standards of fetal growth have been proposed by the Intergrowth project.Reference Villar, Cheikh Ismail and Victora 9 , Reference Villar, Giuliani and Fenton 10

Risk factors for IUGR

IUGR can result from a multitude of risk factors, including maternal and fetal causes. Several maternal factors have been identified, such as undernutrition, which notably affects the activity and/or expression of placental nutrient and ion transporters;Reference Rosario, Jansson and Kanai 11 Reference Settle, Sibley and Doughty 13 chronic diseases, such as preeclampsia;Reference Chaiworapongsa, Chaemsaithong, Yeo and Romero 14 bacterial infection during pregnancy, particularly with Escherichia coli, group B Streptococcus, Listeria monocytogenes, Treponema pallidum or Trichomonas vaginalis; parasitic diseases, such as malaria; viral infection (e.g. human cytomegalovirus or rubella virus);Reference Adams Waldorf and McAdams 15 , Reference Derricott, Jones and Heazell 16 young age (adolescent pregnancy); and alcohol and/or tobacco consumption.Reference Baba, Wikstrom, Stephansson and Cnattingius 17 In addition, pregnancy-induced HTN, preeclampsia and placental insufficiency are known causes of asymmetrical IUGR (defined as restriction of weight followed by length).Reference Maruyama, Shinozuka and Kondoh 18 Among the fetal causes, chromosomal anomalies (including trisomy of chromosome 13, 18 or 21; tri- and polyploidies; Mulibrey nanism; 3-M, Bloom and Turner syndromes; and Majewski osteodysplastic primordial dwarfism type II)Reference Hall 19 and fetal structural defects, such as congenital heart disease,Reference Malik, Cleves and Zhao 20 result in symmetrical IUGR (defined as global growth restriction), which is usually more severe than asymmetrical IUGR.Reference Maruyama, Shinozuka and Kondoh 18

Along with maternal causes, paternal health has also been identified as a possible contributor to IUGR. Insulin resistance, smoking habits, elevated blood pressure, endothelial dysfunction, upper body fat distribution and an atherogenic lipid profile have all been suggested as potential paternally determined factors that correlate with IUGR.Reference Hillman, Peebles and Williams 21 These factors presumably impact fetal growth through epigenetic processes.Reference Li, Tsuprykov, Yang and Hocher 22

IUGR is now considered a critical public health issue because of its high perinatal mortality rate and long-term consequences. As numerous epidemiological studies have reported, infants born with fetal growth restriction have an increased risk of developing non-communicable chronic diseases, notably cardiovascular (e.g. systemic HTN and coronary artery disease) and renal [chronic kidney disease (CKD)] diseases, later in life. These observations are consistent with the concept of Developmental Origins of Health and Disease, which suggests that conditions affecting specific sensitive developmental periods, from conception throughout pregnancy to early infancy, ‘program’ tissue/organ structure and function throughout life in a process known as developmental plasticity that is adapted to short term, prevailing environmental conditions but possibly not to the further life course. The underlying mechanisms are not clearly defined. In parallel with the long-term effects of decreased nephron numbers and hyperactivity of the hypothalamic–pituitary–adrenal axis in these infants, endothelial dysfunction may also contribute to the development of certain chronic diseases in adulthood.

Endothelium dysfunction in individuals born with IUGR

The endothelium: a major role in vascular homeostasis

The endothelium plays a major role in maintaining vascular homeostasis and is one of the largest organs in the human body, consisting of more than 1014 cells lining the vascular network. It is intimately involved in the balance between vasodilation and vasoconstriction and between thrombogenesis and fibrinolysis, the inhibition and promotion of smooth muscle cell proliferation and migration, and the prevention and stimulation of platelet adhesion and aggregation.Reference Minshall, Tiruppathi, Vogel and Malik 23 All these functions are mediated by the release of numerous vasoactive factors, such as nitric oxide (NO) and endothelin. In this respect, the maintenance of endothelial structural and functional integrity is essential for vascular homeostasis; therefore, impaired endothelial function can lead to the development of vascular-related diseases.

Convincing evidence suggests that endothelial dysfunction during early childhood and persisting to adulthood in individuals born with IUGR is a key event in the development of HTN, atherosclerosis, coronary heart disease and CKD later in life. In these individuals, endothelial dysfunction primarily manifests as impaired endothelium-dependent vasodilation and vascular remodeling.

Impaired endothelium-dependent vasodilation in individuals born with fetal growth restriction

Endothelium-dependent vasodilation can be clinically evaluated using flow-mediated brachial artery tests, plethysmography or skin perfusion in response to acetylcholine using the laser Doppler technique.Reference Purnomowati, Kariadi, Achmad, Mose and Setianto 24 Reference Martin, Lindblad and Norman 26 Impaired endothelium-dependent vasodilation has been described in children (9–11 years)Reference Leeson, Thorne and Donald 27 Reference Goodfellow, Bellamy and Gorman 29 and young adults (20–28 years) born with fetal growth restrictionReference Leeson, Kattenhorn, Morley, Lucas and Deanfield 30 and in umbilical and placental vessels derived from growth-restricted fetuses.Reference Krause, Carrasco-Wong and Caniuguir 31

An association between fetal growth restriction and impaired NO-dependent vasorelaxation has also been observed in several animal models, mainly in rats, mice and sheep. IUGR can be induced in rats by exposure to a maternal low-protein diet (LPD, containing 9% casein)Reference Yzydorczyk, Gobeil and Cambonie 32 or restricted diet (50% of normal intake) and in sheep by single in utero umbilical artery ligation (at 105–110 days gestation); these diets and procedures result in low birth weight (LBW) offspring and lead to impaired endothelium-dependent vasodilation in small arteries,Reference Pladys, Sennlaub and Brault 33 , Reference Brawley, Itoh and Torrens 34 the aortaReference Franco, Arruda and Dantas 35 and coronary arteriesReference Tare, Parkington and Wallace 36 in adulthood.

The effects on endothelium-dependent vasodilation are more pronounced in males, whereas females seem to be protected by the NO-dependent vasoprotective role of estrogens. However, impaired endothelium-dependent vasodilation has been observed in female Wistar rats born with IUGR stemming from maternal undernutrition.Reference Franco, Arruda and Dantas 35 As suggested by Borwick et al.,Reference Borwick, Rhind, McMillen and Racey 37 fetal undernutrition may decrease estrogen synthesis, ultimately leading to ovarian damage. Interestingly, estrogen-mediated vasoprotective activity has been reported in humans; specifically, postmenopausal women taking conjugated equine estrogens (0.625 mg for 28 days) showed improved vascular NO-dependent relaxation of the brachial artery.Reference Hurtado, Celani and Geber 38

Vascular remodeling in individuals born with fetal growth restriction

Endothelial activation

Endothelial dysfunction is associated with leukocyte infiltration and the adhesion of monocytes, macrophages and low-density lipoprotein (LDL), which is oxidized to OxLDL in the arterial wall. This leads to foam cell formation and initiates atherogenesis. In addition, monocytes and macrophages secrete higher levels of cytokines and pro-inflammatory proteins such as interleukin-6 (IL-6), tumor necrosis factor-α and C-reactive protein (CRP).Reference Gleeson, Bishop and Stensel 39 These events create a vicious cycle: neutrophils and macrophages produce higher levels of IL-6 in response to inflammation, which in turn increases CRP production in the liver. CRP decreases NO availability and increases endothelin-1 production, thereby contributing to impaired endothelium-dependent vasodilation and leading to irreversible vascular damage.Reference Gleeson, Bishop and Stensel 39 , Reference Leinonen, Hurt-Camejo and Wiklund 40 Elevated levels of pro-inflammatory markers and endothelial activators are characteristic of middle-aged adults (45–64 years) born with LBW, indicating that endothelial dysfunction is patent in these individuals.Reference Pellanda, Duncan and Vigo 41

Vascular structural changes

Histopathological analyses showed that the first atherosclerotic lesions begin to develop in the abdominal aorta.Reference Teeninga, Schreuder, Bokenkamp, Delemarre-van de Waal and van Wijk 42 Increased arterial wall thickness, measured using non-invasive assessments of the intima-media or carotid intima-media thickness, has been observed in newbornsReference Skilton, Evans, Griffiths, Harmer and Celermajer 43 Reference Cosmi, Visentin, Fanelli, Mautone and Zanardo 46 and young childrenReference Crispi, Figueras and Cruz-Lemini 47 , Reference Crispi, Bijnens and Figueras 48 and persists in adults (27–30 years) born after IUGR, and it is particularly apparent in those with exaggerated postnatal growth.Reference Oren, Vos and Uiterwaal 49

Hypoxia and oxidative stress in vascular remodeling

Hypoxia and oxidative stress have been implicated in vascular remodeling.

Placental insufficiency is related to reduced nutrient and oxygen delivery to the fetus, contributing to the development of fetal growth restriction. Several maternal factors, such as living at a high altitude, HTN, anemia, pulmonary disease, preeclampsia, drugs and/or tobacco consumption can contribute to fetal hypoxia,Reference Jensen and Moore 50 which can induce IUGR, LBWReference Lueder, Kim, Buroker, Bangalore and Ogata 51 and increase the risk of cardiovascular diseases (CVD) later in life.Reference Barker 52 , Reference Barker, Osmond, Golding, Kuh and Wadsworth 53 During fetal development, hypoxia plays a crucial role by driving vasculogenesis/angiogenesis, hematopoeisis and chondrogenesis.Reference Giaccia, Simon and Johnson 54 However, prolonged in utero hypoxia can lead to detrimental effects. In growth-restricted fetuses, circulating levels of angiopoietin-2, an angiogenic factor up-regulated by hypoxia, were increased at postnatal day 4 compared with appropriate-for-gestational age infants, thus contributing to postnatal vascular remodeling.Reference Malamitsi-Puchner, Boutsikou and Economou 55

Oxidative stress can be defined by decreased antioxidant defenses and increased reactive oxygen species (ROS) production. Under physiological conditions, ROS play an important role as a regulator of vascular functions such as migration, growth, smooth muscle and endothelial cell survival, and the secretion of extracellular matrix proteins. However, uncontrolled ROS production can contribute to vascular diseases.Reference Griendling and Harrison 56 , Reference Irani 57 ROS have been implicated in the hypertrophy and hyperplasia of vascular smooth muscle cells. In vascular cells (endothelial cells and vascular smooth muscle cells, adventitial fibroblasts), the main enzyme responsible for ROS production is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase.Reference Touyz and Schiffrin 58

Angiotensin II (AngII), via Angiotensin type 1 receptor (AT1R), has been implicated in increasing superoxide anion levels followed by increased hydrogen peroxide production, which induces long-term outcomes of AngII, such as hypertrophy and hyperplasia of vascular smooth muscle cells.Reference Ushio-Fukai, Zafari, Fukui, Ishizaka and Griendling 59 , Reference Griendling, Minieri, Ollerenshaw and Alexander 60 The flavoprotein inhibitor diphenyleneiodonium (DPI)Reference Griendling, Minieri, Ollerenshaw and Alexander 60 and catalase overexpressionReference Zafari, Ushio-Fukai and Akers 61 inhibit these vascular defects. In a rat model of IUGR induced by maternal LPD associated with adult HTN, we observed increased ex vivo vasoreactivity of the carotid rings to AngII, mediated by AT1R, which was normalized by DPI and apocynin (NADPH oxidase inhibitor) pre-incubation.Reference Yzydorczyk, Gobeil and Cambonie 32

The regulation of extracellular matrix proteins such as collagen and elastin can be modulated by ROS. Elastinolysis and collagenolysis play crucial roles in arterial remodeling and vascular diseases.Reference Tyagi and Simon 62 Metalloproteinases (MMPs) and their related tissue inhibitors of metalloproteinases are enzymes secreted by macrophages and vascular smooth muscle cells. MMP-2 and MMP-9 cleave gelatin, collagen and elastin, and have been associated with vascular diseases.Reference Chow, Cena and Schulz 63 ROS have been demonstrated to activate MMPs.Reference Rajagopalan, Meng, Ramasamy, Harrison and Galis 64 Increased circulating levels of MMP-2 and MMP-9 and increased MMP-2/TIMP-2 and MMP-9/TIMP-2 ratios have been observed in children who were small for gestational age and are positively correlated with systolic blood pressure and vascular function.Reference Sesso and Franco 65 In a developmental programming animal model of HTN induced by neonatal oxygen exposure, we observed increased aortic MMP-2 and TIMP-1 and reduced TIMP-2 staining as early as 4 weeks of age, indicating a shift in the balance toward degradation of the extracellular matrix and increased collagen deposition.Reference Huyard, Yzydorczyk and Castro 66 These data suggest that early changes could contribute to the onset of the elevated blood pressure and arterial stiffness observed at adulthood in this animal model.Reference Yzydorczyk, Comte and Cambonie 67 , Reference Mivelaz, Barbier, Cloutier, Fouron and Nuyt 68

Mechanisms involved in endothelial dysfunction in individuals born with fetal growth restriction

Impaired NO bioavailability

The endothelium-mediated release of NO is widely accepted as the key determinant of endothelial function, and reduced NO bioavailability has been linked to most serious vascular pathologies.Reference Chatterjee, Black and Catravas 69 In particular, the loss of NO production contributes to impaired endothelium-dependent vasodilation and to endothelium activation by improving the recruitment of pro-inflammatory cytokines, such as vascular cell adhesion molecule-1 and ICAM-1, and the infiltration of leukocytes into the vessel wall.Reference Förstermann and Münzel 70 Reference De Caterina, Libby and Peng 72 In normal pregnancies, NO synthesis is up-regulated, as reflected by increased nitrite/nitrate concentrations in maternal and fetal circulation, thus mediating maternal cardiovascular adaptations and the low systemic and umbilical vascular resistance in the fetus. In pregnancies complicated by IUGR, research findings are inconsistent. Some have displayed a decrease in NO metabolite concentrations in maternal and/or fetal serum, reflecting reduced NO synthesis compared with controls.Reference Hata, Hashimoto and Manabe 73 , Reference Singh, Singh and Sharma 74 Other have found higher nitrite/nitrate concentrations in umbilical venous plasmaReference Malamitsi-Puchner, Boutsikou and Economou 55 or an increase in endothelial nitric oxide synthase (eNOS) protein staining in placental vessels compared with normal pregnancies, suggesting that increased NO production could be a compensatory response to improve blood flow in the placenta.Reference Lyall, Greer, Young and Myatt 75 , Reference Myatt, Eis, Brockman, Greer and Lyall 76

Decreased NO synthesis, evaluated in terms of nitrate/nitrite production, was observed in animal models of IUGR induced by a reduction in utero placental perfusion pressureReference Payne, Alexander and Khalil 77 or maternal LPDReference Sathishkumar, Elkins, Yallampalli and Yallampalli 78 , Reference Bourdon, Parnet and Nowak 79 and in a rat model of developmental programming of HTN induced by exposing pregnant rats to androgens.Reference Sathishkumar, Elkins, Yallampalli, Balakrishnan and Yallampalli 80

Reduced NO bioavailability may result either from altered NO synthesis or from NO scavenging by other molecules, such as ROS.

Impaired eNOS functionality

Under physiological conditions, NO is synthetized in the vasculature by eNOS, using L-arginine (L-Arg) as a substrate and tetrahydrobiopterin (BH4) as a cofactor. There are contradictory data on eNOS expression in individuals born after IUGR. In humans, independent studies have indicated that eNOS expression is increased in the umbilical arteries of babies born after fetal growth restriction, suggesting that activated NO synthesis may be a compensatory mechanism to improve placental blood flow.Reference Krause, Carrasco-Wong and Caniuguir 31 , Reference Hracsko, Hermesz and Ferencz 81 However, these results are controversial because they could not be replicated.Reference Dellee, Tobias, Li and Mildenberger 82 Moreover, differences in eNOS expression have been observed in human endothelial cells isolated from the umbilical arteries (HUAECs) or veins (HUVECs) of IUGR newborns. eNOS expression is increased in IUGR-HUAECs but decreased in IUGR-HUVECs. These differences may be explained by the type of vessel (artery v. vein) or could be the consequence of altered blood flow and oxygen levels in pregnancies complicated by IUGR.Reference Maruyama, Shinozuka and Kondoh 18

In animal studies, eNOS expression varies depending on the animal model of IUGR used. In Dahl-S rats fed a high-salt diet to induce fetal growth restriction, the placental eNOS messenger RNA (mRNA) expression level was significantly increased compared with controls.Reference Takushima, Nishi and Nonoshita 83 In an animal model of IUGR induced by placental insufficiency using hyperthermic exposure, placental and umbilical artery eNOS protein in the placenta was decreased at mid-gestation but increased near term.Reference Arroyo, Anthony, Parker and Galan 84

However, eNOS expression and activity and the gender effect seem particularly sensitive to undernutrition. In fact, decreased eNOS expression and/or activity have been reported in animal models of IUGR induced by intrauterine undernourishment.Reference Franco, Arruda and Dantas 35 In a rat IUGR model induced by intrauterine undernourishment,Reference Franco, Arruda and Dantas 35 eNOS expression was decreased only in males, whereas eNOS activity was decreased in both males and females. This reduction in eNOS activity in females, which is probably the consequence of decreased estrogen levels, could explain the impaired endothelium-dependent vasodilation observed in this animal model.Reference Franco, Arruda and Dantas 35 The modulation of eNOS activity by estrogens has been confirmed in vitro. Long-term estrogen treatment of cultured human and bovine endothelial cells up-regulates eNOS activity.Reference Tolbert, Thompson, Bouchard and Oparil 85

Upregulation of the arginase pathway

Arginases produce urea and ornithine, using L-Arg as a substrate. By competing with eNOS for the bioavailability of L-Arg, arginases can indirectly contribute the reduction of NO synthesis by eNOS. Accordingly, arginase up-regulation is an important factor that drives endothelial dysfunction. Increased arginase-2 expression was observed in human umbilical endothelium from IUGR fetuses.Reference Krause, Costello and Munoz-Urrutia 86 Pre-incubation with S-(2-boronoethyl)-L-cysteine, an arginase inhibitor, improved ex vivo endothelium-dependent relaxation in umbilical and placental vessels from babies born after fetal growth restrictionReference Krause, Carrasco-Wong and Caniuguir 31 and in aortic rings from a rat IUGR model induced by maternal LPD (personal unpublished data). These data suggest that arginase activity was increased in these vessels.

Increased asymmetric dimethylarginine (ADMA) levels

ADMA, an endogenous NO synthase inhibitor, is also considered an early marker and mediator of endothelial dysfunction. ADMA acts as a competitor of L-Arg, thereby inhibiting NO synthesis by eNOS. However, the observations in human studies are controversial. In pregnancies complicated by IUGR, ADMA levels in maternal serum were found to be either increasedReference Laskowska, Laskowska and Oleszczuk 87 , Reference Gumus, Atalay, Cetinkaya Demir and Sahin Gunes 88 or decreased compared with those in normal gestations during the first (11–14 weeks), second (20–24 weeks) and third trimesters (28–35 weeks).Reference Rizos, Eleftheriades and Batakis 89 Estrogen therapy could improve endothelial function by reducing ADMA levels. Clinical data revealed that estrogen therapy, chiefly the oral form, decreased plasma ADMA concentrations and therefore improved NO production in healthy postmenopausal women.Reference Post, Verhoeven and van der Mooren 90 , Reference Karkanaki, Vavilis, Traianos, Kalogiannidis and Panidis 91

In animal models of atherosclerosis (rabbits and monkeys), endothelial dysfunction was associated with increased ADMA levels.Reference Yu, Li and Xiong 92 , Reference Boger, Bode-Boger, Sydow, Heistad and Lentz 93 To the best of our knowledge, ADMA levels have not been assessed in animal models of IUGR.

Oxidative stress

Oxidative stress plays an important role in endothelial dysfunction. ROS, particularly the superoxide anion (O2 ), play a central role in vascular physiology, and their overproduction is especially relevant to vascular pathologies.Reference Griendling and FitzGerald 94 In IUGR placentae, markers of oxidative stress, such as 8-hydroxy-2’-deoxyguanosine, redox factor-1,Reference Burton and Jauniaux 95 , Reference Takagi, Nikaido and Toki 96 malondialdehyde and oxidized LDL, are increased in venous cord blood.Reference Maisonneuve, Delvin and Edgard 97 Therefore, it has been suggested that oxidative stress is involved in both the short- and long-term modulation of endothelial function in individuals born after IUGR.Reference Webster, Roberts and Myatt 98 Oxidative stress affects the NO pathway by influencing NO synthesis and bioavailability. NO rapidly reacts with O2 to form peroxynitrite, a highly reactive and toxic species, which reduces endothelium-dependent relaxationReference Kossenjans, Eis, Sahay, Brockman and Myatt 99 and accelerates the development of pre-atherosclerotic lesions.Reference Santilli, D’Ardes and Davi 100

As mentioned above, L-Arg and BH4 are crucial for NO production. A deficit in substrate and/or cofactor leads to enzymatic uncoupling, which causes eNOS to produce O2 rather than NO,Reference Yzydorczyk, Comte and Huyard 101 thus contributing to endothelial dysfunction and impaired endothelium-dependent vasodilation.Reference Vasquez-Vivar, Kalyanaraman and Martasek 102 , Reference d’Uscio, Santhanam and Katusic 103 Decreased BH4 bioavailability can contribute to eNOS uncoupling. Regardless of whether the BH4 level is sufficient, the oxidation of L-Arg is coupled with the reduction of oxygen molecules to form L-citrulline and NO. However, BH4 bioavailability can be decreased through reduced production,Reference Yang, Huang, Kaley and Sun 104 increased oxidationReference Landmesser, Dikalov and Price 105 or impaired recycling of the oxidized form (BH2)Reference Chalupsky and Cai 106 therefore leading to eNOS uncoupling.

Increased O2 production up-regulates ADMA levels, thus worsening endothelial dysfunction.Reference Sydow and Munzel 107 In humans, impaired NO-dependent vasodilation in placental vessels from IUGR pregnancies is coupled with a higher sensitivity to oxidative stress.Reference Schneider, Hernandez and Farias 108

Increased O2 production mediated by NADPH oxidase and eNOS uncoupling was associated with defective endothelial function in a rat model of HTN induced by deoxycorticosterone and saline treatmentReference Mitchell, Cook, Danchuk and Puschett 109 and in a rat model of IUGR induced by maternal diet restriction (50% of ad libitum intake throughout gestation) or LPD (9% casein).Reference Yzydorczyk, Gobeil and Cambonie 32 , Reference Oliveira, Akamine and Carvalho 110

Endothelial progenitor cells (EPCs) dysfunction

Endothelial dysfunction is characterized by impaired vasculogenesis and decreased repair capacity, functions that are mediated by circulating EPCs. These cells are bone marrow-derived stem cells that can differentiate into mature endothelial cells, thus contributing to postnatal vasculogenesis and endothelial repair at damage sites.Reference Asahara, Murohara and Sullivan 111 EPCs subsets are differentiated by their phenotype and functional properties. The myeloid subset represents early EPCs, called colony-forming unit-endothelial cells, that appear early in cultures and display endothelial markers but do not form vessels in vivo.Reference Yoder, Mead and Prater 112 , Reference Purhonen, Palm and Rossi 113 Endothelial colony-forming cells (ECFCs), the true angioblasts, appear later and display properties such as proliferation, auto-renewal, migration and differentiation; additionally, they can support vascular growth and neovascularization. Both loss and impaired function of EPCs have been identified as markers of endothelial dysfunction, as described by Hill et al. Reference Hill, Zalos and Halcox 114 In adult men with different degrees of cardiovascular risk but without a history of cardiovascular disease, levels of circulating EPCs have been identified as a surrogate biological marker of vascular function and cumulative cardiovascular risk.Reference Hill, Zalos and Halcox 114 In pregnancy-related complications, most notably IUGR, aberrant vasculature and abnormal endothelial function were found on both the maternal and fetal sides of the placenta, and it is believed that altered fetal circulating EPCs contribute to these complications.Reference Sipos, Crocker, Hubel and Baker 115 We and others have evaluated ECFCs isolated from LBW newborns and observed altered angiogenic properties in vitro, as evidenced by decreased numbers of colonies and sprouts,Reference Hwang, Kwon and Kwon 116 and in vivo, as shown by a reduction in the number of perfused vessels.Reference Ligi, Simoncini and Tellier 117 Moreover, an imbalance between angiogenic and anti-angiogenic factors was noted.Reference Ligi, Simoncini and Tellier 117 , Reference Ligi, Simoncini and Tellier 118 These data suggest that the impairment of early angiogenic properties (structural and functional) could predispose LBW infants to endothelial dysfunction later in life.

Vascular senescence

Vascular senescence can contribute to endothelial dysfunction.Reference Minamino and Komuro 119 It is characterized by a state of irreversible (replicative senescence) or reversible (stress-induced senescence) growth arrest, the expression of negative cell cycle regulators (such as p53 and p16) and increased senescence-related β-galactosidase staining.Reference Erusalimsky and Fenton 120 Senescent endothelial cells have a decreased ability to form new vascular structures; therefore, they contribute to impaired endothelial function. Sirtuins (SIRTs), particularly SIRT1, belong to a family of proteins involved in the regulation of many cellular processes, including senescence. SIRT1 is highly expressed in endothelial cells, wherein it regulates numerous functions, such as NOS expression and cellular senescence.Reference Borradaile and Pickering 121 The depletion of SIRT1 expression in endothelial cells led to endothelial dysfunction and premature senescence in several models of cardiovascular diseases, whereas overexpression of SIRT1 protected endothelial cells from senescence-associated morphological and molecular changes.Reference Ota, Akishita and Eto 122 ECFCs from LBW newborns exhibit stress-induced vascular senescence characterized by growth arrest, increased β-galactosidase activity, and p16INK4a expression, all of which are mediated by decreased SIRT1 levels.Reference Vassallo, Simoncini and Ligi 123 Therefore, stress-induced vascular senescence is coincident with impaired angiogenic properties and could participate in the endothelial dysfunction observed later in life in individuals born after IUGR.

Relationship between IUGR and cardiovascular and renal outcomes later in life

Early endothelial dysfunction observed in individuals born after IUGR could persist for the long term and lead to the onset of cardiovascular-related diseases.

Systemic HTN

Epidemiological studies have highlighted an inverse correlation between LBW and increased blood pressure in infancy,Reference Fattal-Valevski, Bernheim and Leitner 124 adolescence,Reference Rossi, Tauzin and Marchand 125 , Reference Chiolero, Cachat, Burnier, Paccaud and Bovet 126 young adulthoodReference Leon, Johansson and Rasmussen 127 , Reference Nilsson, Ostergren, Nyberg, Soderstrom and Allebeck 128 and adulthood.Reference Gennser, Rymark and Isberg 129 Reference Law and Shiell 132 Some authors have questioned these results, suggesting that the data were inappropriately adjusted for confounding factorsReference Huxley, Neil and Collins 133 that could potentially damage kidneys and/or vascular endothelial cells early in life (e.g. nephrotoxic drugs or umbilical catheter placement). Recent data have indicated that the risk of HTN is not only linked to birth weight but is also amplified by postnatal overfeeding, leading to exaggerated catch-up growth.Reference Tauzin, Rossi and Grosse 134

Several animal models have shown that IUGR induced by ligation of the bilateral uterine vessels, prenatal exposure to hypoxia (11.5 v. 21% O2) or glucocorticoids, maternal global undernutrition, caloric restriction or LPD during gestation induces HTN in adulthoodReference Yzydorczyk, Gobeil and Cambonie 32 , Reference Wlodek, Westcott, Siebel, Owens and Moritz 135 Reference Gilbert, Lang, Grant and Nijland 141 and is often associated with vascular dysfunction.Reference Yzydorczyk, Gobeil and Cambonie 32 , Reference Ozaki, Nishina, Hanson and Poston 142 , Reference Cambonie, Comte and Yzydorczyk 143 However, it is not well established whether HTN precedes endothelial dysfunction. Some clinical investigations have suggested that endothelial dysfunction is a primary defect in essential HTN that appears before the increase in blood pressure,Reference Taddei, Virdis, Mattei, Arzilli and Salvetti 144 but other observations have hinted that endothelial dysfunction is a consequence of elevated blood pressure. Different animal models of HTN induced by aortic coarctation (rabbits),Reference Miller, Pinto and Mullane 145 a high-salt diet (rats)Reference d’Uscio, Barton, Shaw, Moreau and Luscher 146 or neonatal hyperoxia (rats)Reference Yzydorczyk, Comte and Cambonie 67 showed selective impairment of endothelium-dependent vasodilation secondary to increased blood pressure. However, in an animal model of IUGR caused by maternal LPD during gestation, impaired endothelium-dependent relaxation preceded the onset of increased blood pressure (personal unpublished data).

Coronary heart disease

Impaired endothelial function plays a major role in the development and progression of atherosclerosis,Reference Verma and Anderson 147 , Reference Ludmer, Selwyn and Shook 148 which ultimately leads to coronary heart disease. Many studies have proposed a relationship between birth weight and coronary heart disease: some showed an inverse relationship between LBW and increased risk of coronary heart disease,Reference Barker, Gluckman and Godfrey 149 Reference Wang, Shu and Sheng 151 whereas others found no significant correlationReference Eriksson, Tibblin and Cnattingius 152 or a positive correlation only in males.Reference Banci, Saccucci and Dofcaci 153 Interestingly, the risk of coronary heart disease decreases with increasing birth weight. In fact, a 1-kg increase in birth weight was associated with a 10–20% decreased risk of coronary heart disease later in life.Reference Wang, Shu and Sheng 151

CKD

The role of vascular components in the renal system is of particular significance because the kidneys receive ~20–25% of the total cardiac output. However, the contribution of the endothelial compartment to kidney development has been the subject of many hypotheses. Previous experiments showed that a significant proportion of the renal endothelium is derived from a resident precursor, the metanephric mesenchyme.Reference Abrahamson, Robert, Hyink, St John and Daniel 154 Sprouting angiogenesis from the major renal vessels plays a significant role in forming the kidney endothelium, thus giving rise to most of the renal vessels and glomerular capillaries.Reference Hyink, Tucker and St John 155 Endothelial dysfunction is involved in the development and progression of CKD.Reference Stehouwer, Henry and Dekker 156 Patients with CKD display microalbuminuria, which is thought to reflect endothelial damage in the capillary system of the renal medulla and increased endothelial permeability.Reference Stehouwer, Henry and Dekker 156 Reference Perticone, Maio and Perticone 159 Capillary damage is characterized by increased plasma concentrations of endothelium-derived proteins, such as von Willebrand factor, tissue-type plasminogen activator and urokinase-type plasminogen activator, and increased concentrations of markers of endothelial cell injury, such as soluble thrombomodulin. Decreased endothelium-dependent vasodilation occurs in end-stage kidney disease.Reference Gris, Branger and Vecina 160 Several epidemiological and experimental studies have shown that intrauterine insults are associated with the development of CKD. In humans, birth weight is positively correlated with glomerular number and inversely correlated with glomerular volume.Reference Manalich, Reyes, Herrera, Melendi and Fundora 161 In a meta-analysis of 18 studies, infants born after fetal growth restriction appeared to have a significantly higher risk of albuminuria [odds ratio (OR), 1.81; 95% confidence interval (CI), 1.19–2.77], end-stage renal disease (OR, 1.58; 95% CI, 1.33–1.88), or a low estimated glomerular filtration rate (OR, 1.79; 95% CI, 1.31–2.45).Reference White, Perkovic and Cass 162 Similar to HTN, the impairment of glomerular and tubular function secondary to IUGR is further amplified by environmental insults, such as drug exposure during the neonatal periodReference Giapros, Papadimitriou, Challa and Andronikou 163 or overweight in adulthood.Reference Silverwood, Pierce and Hardy 164

Several animal models have enabled the identification of mechanisms involved in the development of renal dysfunction later in life. Rat models of IUGR induced by exposure to maternal LPD followed by early postnatal overnutrition during the lactation period or not according litter size reduction or increased protein intake to induce accelerated postnatal growth displayed alterations in renal structural development and a risk of chronic renal failure later in life.Reference Vehaskari, Aviles and Manning 165 Reference Boubred, Daniel and Buffat 169 Decreased glomerular number potentially leads to reduced filtration capacity, reduced salt and water retention and the subsequent development of HTN. Furthermore, early loss of nephron numbers/mass may result in a state of hyperfiltration in the remaining nephrons, which will lead to focal segmental glomerulosclerosis and further loss of glomeruli, thus initiating a vicious circle.Reference Anderson, King and Brenner 170 However, it is not clear whether endothelial dysfunction precedes or is a consequence of CKD. Regarding the impact of postnatal nutrition on renal maturation, rodent models could likely be more affected than humans because nephrogenesis is completed at ~36 weeks of gestation in humans, whereas in rats, this process is completed during postnatal life (between 7 and 10 days of life).

Potential preventive approaches

Several interventions have been identified to potentially prevent IUGR, improve endothelial function and thus antagonize the development of detrimental cardiovascular issues.

Breastfeeding

Breast milk could represent a promising approach, and the easiest one, for improving endothelial function in offspring. In fact, breastfeeding, as opposed to feeding with commercial infant formulas, is one of the best approaches for fighting neonatal oxidative stress because of breast milk’s ability to ‘trap’ free radicals. Breast milk contains enzymatic and non-enzymatic components such as superoxide dismutase, glutathione peroxidase, vitamins (A, C and E), α-carotene, lactoferrin and trace amounts of iron. Breastfeeding could improve endothelial function, primarily due to the presence of lactoferrin, an iron-binding glycoprotein with antioxidant, anti-inflammatory, pro-angiogenic and NO-dependent vasodilator properties. Daily treatment with lactoferrin after unilateral hind limb surgery-induced ischemia in C57BL/6J mice promoted angiogenesis, activated endothelial function via an NO-dependent mechanismReference Ikeda, Tajima and Izawa-Ishizawa 171 and protected HUVECs against hydrogen peroxide-induced oxidative stress.Reference Safaeian, Javanmard, Mollanoori and Dana 172

Folate supplementation

Epidemiological studies have shown that folate deficiency is associated with increased cardiovascular risk.Reference Verhaar, Stroes and Rabelink 173 , Reference Robinson, Arheart and Refsum 174 Because of the homocysteine-lowering and antioxidant effects of folate and its ability to modulate eNOS activity and cofactor availability, folic acid supplementation could improve vascular endothelial structure and function.

In a study including patients with coronary heart disease, the circulating form of folic acid, 5-methyltetrahydrofolate, increased NO-dependent vasodilation, reduced vascular superoxide production, and improved enzymatic coupling of eNOS by increasing the availability of BH4.Reference Antoniades, Shirodaria and Warrick 175 Folate supplementation in patients with acute ischemic strokeReference Xia, Li and Wang 176 or HTNReference Wu, Wang and Li 177 decreased plasma ADMA levels, suggesting that folate intake may also be beneficial in these contexts. Moreover, folic acid supplementation during pregnancy increased the birth weight of newborns.Reference Li, Qu and Dang 178

Folate deficiency in ApoE−/− mice was associated with the development of atherosclerotic lesions, which can be prevented by folate supplementation.Reference Alessio, Santos and Debbas 179 Moreover, folate supplementation of a maternal LPD diet prevented the development of increased blood pressure and restored endothelium-dependent vasodilation and eNOS mRNA expressionReference Torrens, Brawley and Anthony 180 and enzyme activity.Reference Stroes, van Faassen and Yo 181

Vitamin supplementation

Studies of animal models of IUGR and developmental programming of CVD have demonstrated that maternal diet supplementation with vitamins C and E can prevent adverse perinatal and long-term outcomes. In an animal model of IUGR induced by high maternal cholesterol levels during the early stages of gestation, maternal dietary supplementation with vitamin E was found to prevent growth restriction in fetuses. Vitamin E has been shown to regulate molecular pathways controlling cell proliferation and viabilityReference Zingg and Azzi 182 and to increase the release of vasodilator prostanoids from human aortic endothelial cellsReference Wu, Liu, Meydani and Meydani 183 and HUVECs,Reference Tran and Chan 184 thus improving placenta–fetal blood flow and thereby increasing nutrient delivery to the fetus.

Vitamin C was found to protect chick embryos against the developmental toxicity of ethanol. Indeed, concomitant injection of vitamin C and ethanol in chick embryos prevented the decreased survival, growth retardation and malformations induced by ethanol alone.Reference Memon and Pratten 185

However, in human studies, these treatments have failed to show clear benefits in terms of birth weight and associated long-term diseases.Reference Hovdenak and Haram 186 Reference Rumbold, Crowther and Haslam 189 A possible explanation is the potential confounding effects of maternal endogenous antioxidant defenses and redox status and maternal vitamin intake resulting from diversified nutrition. Differences in vitamin metabolism between humans and animals could also be involved in the discrepancy between human and animal studies.

Antioxidant therapy

Supplementation with resveratrol, a polyphenolic molecule found at high concentrations in red grapes, berries and peanuts, has been identified as a potential therapeutic strategy for the treatment of cardiovascular diseases, primarily due to its antioxidant properties and ability to modulate the NO signaling pathway. In spontaneously hypertensive rats, maternal dietary supplementation with resveratrol during the perinatal period prevented the onset of HTN in adult offspring.Reference Care, Sung and Panahi 190 Resveratrol also modulates SIRT1 expression. Pre-incubation with resveratrol restored angiogenic capacity and reversed the accelerated senescence of ECFCs from LBW newborns.Reference Vassallo, Simoncini and Ligi 123

Lazaroid is a potent inhibitor of free radical formation, notably O2 -mediated lipid peroxidation. Treatment with Lazaroid reversed HTN in several rat models,Reference Vaziri, Ding, Ni and Gonick 191 , Reference Vaziri, Ni, Oveisi and Trnavsky-Hobbs 192 and the addition of Lazaroid to a maternal LPD diet throughout gestation increased birth weight and reversed later vascular dysfunction in offspring by decreasing oxidative stress.Reference Cambonie, Comte and Yzydorczyk 143

In a guinea pig model of IUGR induced by progressive uterine artery occlusion starting at mid-gestation, maternal N-acetylcysteine treatment during the second half of gestation restored ex vivo eNOS-dependent relaxation in the fetal aorta and umbilical artery and normalized eNOS expression in fetal and umbilical endothelial cells.Reference Herrera, Cifuentes-Zuniga and Figueroa 193

Melatonin, a hormone with antioxidant and anti-inflammatory properties, is involved in regulating circadian and circannual rhythmsReference Hardeland, Cardinali and Srinivasan 194 and could improve endothelial function. Melatonin exhibits direct scavenging activity on O2 ,Reference Tare, Parkington and Wallace 36 , Reference Reiter, Tan, Terron, Flores and Czarnocki 195 , Reference Franco Mdo, Akamine and Aparecida de Oliveira 196 up-regulates antioxidant enzymes such as superoxide dismutase and glutathione peroxidase, and down-regulates pro-oxidant enzymes such as lipoxygenase,Reference Galano, Tan and Reiter 197 thereby increasing NO production and improving its availability to induce vasodilationReference Lopez, Garcia and Escames 198 in different vascular beds.Reference Herrera, Macchiavello and Montt 199 Reference Das, Balonan, Ballard and Ho 202

L-citrulline supplementation

L-citrulline is a precursor of L-Arg. L-citrulline is a non-protein amino acid, which is absent from the regular diet, escapes liver metabolism, has high bioavailability, and is quantitatively converted to arginine in vivo.Reference Curis, Nicolis and Moinard 203 Data have suggested that L-citrulline supplementation improves fetal growth in animal model of IUGR induced by in utero exposure to maternal LPD,Reference Bourdon, Parnet and Nowak 79 probably by improving maternal nutritional status and fetal growth through increased NO synthesis as a result of enhanced L-Arg availability in fetal circulation. L-citrulline can also exert a protective role on vascular endothelium. In fact, it has been proposed that L-citrulline supplementation could represent an alternative to L-Arg supplementation to improve vascular function,Reference Romero, Platt, Caldwell and Caldwell 204 , Reference Chien, Lin and Kuo 205 and it attenuated blood pressure in young normotensive men.Reference Figueroa, Trivino, Sanchez-Gonzalez and Vicil 206 In animal models, ex vivo pre-incubation with L-citrulline prevented endothelial dysfunction induced by ADMA in porcine coronary artery; indeed, such incubation favors L-citrulline to L-Arg recycling and the restoration of NO production, as a consequence of eNOS expression and activity up-regulation, the inhibition of superoxide anion production, and activation of the cyclic guanosine monophosphate (cGMP)Reference Xuan, Lun and Zhao 207 Such direct beneficial effects of L-citrulline on endothelium-dependent relaxation suggest that L-citrulline supplementation could be an efficient way to improve endothelial function in individuals born after fetal growth restriction.

Supplementation with L-Arg and NO mediators

It was reported that L-Arg could be administered to increase maternal NO levels to enhance birth weight and decrease neonatal morbidity.Reference Singh, Singh and Sharma 74 More recently, the combined results of 10 small trials showed that L-Arg supplementation can increase the body weight and gestational age at birth of IUGR fetuses.Reference Chen, Gong, Chen, Luo and Zhang 208 However, this study contrasts with others that reported no benefit of L-Arg therapy.Reference Gui, Jia and Niu 209 , Reference Vadillo-Ortega, Perichart-Perera and Espino 210 Such differences could be explained by the different route of administration (oral or intravenous). In fact, with oral administration, 40% of L-Arg is degraded by the small intestine and metabolized by arginase in the liver. Therefore, poor L-Arg availability in the blood could decrease its efficacy.Reference Curis, Nicolis and Moinard 203 , Reference Wu, Bazer and Cudd 211

Among NO modulators, phosphodiesterase inhibitors are promising agents for improving uterine perfusion in pregnancies complicated by IUGR. Type 5 phosphodiesterase (PDE5) is one of the enzymes responsible for the degradation of cGMP to GMP in smooth muscle. Therefore, inhibiting PDE5 delays the breakdown of cGMP and increases vasorelaxation. Sildenafil citrate (Viagra®) is probably the most famous PDE5 inhibitor. In women whose pregnancies were complicated by IUGR, sildenafil citrate improved fetoplacental perfusionReference Dastjerdi, Hosseini and Bayani 212 and decreased the ex vivo vasoconstriction (in response to the thromboxane analogue U46619) of myometrial small arteries.Reference Wareing, Myers, O’Hara and Baker 213

In animal models, parenteral administration of L-Arg (from day 60 of pregnancy to parturition) to underfed ewes prevented fetal growth restriction,Reference Lassala, Bazer and Cudd 214 and in a rat model of IUGR induced by maternal LPD, pre-incubating the aortic rings with L-Arg restored impaired endothelium-dependent vasodilation (personal unpublished data). Sildenafil citrate supplementation reversed the maternal effects of preeclampsia by improving uteroplacental and fetal perfusionReference Herraiz, Pellicer and Serra 215 in a Wistar rat model and increased fetal size in pregnant rats exposed to hypoxia at the end of gestation (18–20 days).Reference Refuerzo, Sokol and Aranda 216

Epigenetic markers of endothelial dysfunction

Epigenetics plays a major role in the developmental origins of health and diseases.Reference Cutfield, Hofman, Mitchell and Morison 217 Epigenetics can be defined as a phenomenon of altered phenotypic expression of heritable genetic information without changes in the DNA sequence. Three main pathways can silence, activate or regulate the level and time of expression of many genes: DNA methylation, histone modifications (acetylation, methylation, ubiquitination, phosphorylation or ADP-ribosylation) and small non-coding RNAs, such as microRNAs (miRNAs).Reference Chen and Zhang 218 , Reference McKay and Mathers 219 In general, these three epigenetic mechanisms appear to work together to regulate gene expression. DNA methylation or histone modifications can alter the expression of miRNAs, which can in turn regulate the epigenetic processes of DNA methylation and histone modifications.

DNA methylation

DNA methylation has been known to be particularly sensitive to an adverse early environment. DNA methylation occurs through the binding of a methyl group in position 5 of the cytosine ring dinucleotide CpG sequences present in the DNA by DNA-methyltransferase, which can methylate and demethylate the DNA, thus making the modification reversible.Reference Kangaspeska, Stride and Metivier 220 In general, low levels of DNA methylation (hypomethylation) are associated with increased gene activity, whereas high levels of methylation (hypermethylation) are associated with gene repression.Reference Lorenzen, Martino and Thum 221 Moreover, hydroxymethylated cytosine [5-hydroxymethylcytosine (5 hmeC)] has been identified as another functional DNA modification, representing an intermediate state of active DNA demethylation and also influencing gene expression.Reference Ito, D’Alessio and Taranova 222 , Reference Kriaucionis and Heintz 223

eNOS expression in HUAECs and HUVECs of IUGR pregnancies can be controlled by DNA methylation levels. eNOS protein and mRNA levels were increased in HUAECs but decreased in HUVECs from IUGR pregnanciesReference Krause, Costello and Munoz-Urrutia 86 and were associated in the eNOS promoter with decreased DNA methylation at CpG −352 in IUGR-HUAECs and an increased in IUGR-HUVECs. In addition, in human umbilical artery endothelial cells from patients with placental insufficiency, levels of 5 hmeC at the eNOS transcription start site directly correlated with elevated eNOS levels.Reference Postberg, Kanders and Forcob 224 In a guinea pig model of IUGR, increased eNOS expression was associated with decreased DNA methylation levels in eNOS promoter of endothelial cells derived from aorta, femoral and umbilical arteries; such modifications were prevented by maternal administration of N-acetylcysteine.Reference Herrera, Cifuentes-Zuniga and Figueroa 193

Histone modifications

In the nucleus, DNA is packaged into chromatin as repeating units of nucleosomes, which form a ‘beads-on-a-string’ structure that can compact into higher order structures to affect gene expression. Nucleosomes are composed of 146-bp DNA wrapped in histone octamers (composed of two H2A, H2B, H3 and H4) and are connected by a linker DNA, which can associate with histone H1 to form heterochromatin. Histone proteins contain a globular domain and an amino-terminal tail, which can be post-translationally modified. The post-translational modification of lysine (acetylation, methylation, ubiquitination, sumoylation), arginine (methylation) and serine and threonine (phosphorylation) are the most commonly described modifications.Reference Chen and Zhang 218 , Reference Canani, Costanzo and Leone 225 In general, the acetylation of histones H3 and H4 is associated with increased gene expression and has been shown to regulate the angiogenic function of endothelial cells.

Levels of H3K9ac and H2A.Zac were significantly higher at the eNOS transcription start site and were directly correlated with elevated eNOS levels observed in the human umbilical artery endothelial cells from patients with placental insufficiency.Reference Postberg, Kanders and Forcob 224 In addition, increased histone H3 acetylation in the endothelin-1 promoter of pulmonary vascular endothelial cells and in the peripheral leucocytes in a IUGR rat model induced by maternal undernutrition has been correlated with higher endothelin-1 expression, which could increase the risk of pulmonary disorders (pulmonary HTN or asthma) later in life.Reference Xu, Xu and Fu 226 Recently, we observed that SIRT1 repression in ECFCs from LBW newborns, associated with premature senescence, could be modulated by changes in ‘active’ or ‘repressive’ epigenetic marks. The ‘active’ marks trimethyl-H3K4 associated with the SIRT1 promoter were significantly decreased in LBW newborns compared with controls, whereas the ‘repressive’ marks trimethyl-H3K9, associated with heterochromatin formation, were increased.Reference Vassallo, Simoncini and Ligi 123

Non-coding RNAs

MiRNAs are small single-strand RNAs that do not encode proteins. Each miRNA binds to specific mRNAs, resulting in the degradation of target mRNA or the inhibition of its translation into protein. miRNAs regulate the post-transcriptional expression level of many genes and processes such as apoptosis, cell growth and differentiation in a large range of tissues,Reference Shruti, Shrey and Vibha 227 , Reference Sayed and Abdellatif 228 and notably in the regulation of endothelial functions. MiR-21 expression is increased in cases of shear stress, which helps to protect endothelial cells by decreasing apoptosis and increasing eNOS expression and NO production.Reference Weber, Baker, Moore and Searles 229 However, in atherosclerotic plaques, an up-regulation of miR-21 decreases the function of superoxide dismutase, which leads to increased ROS production and decreased migration of the progenitor cells.Reference Fleissner, Jazbutyte and Fiedler 230 MiR-221 and miR-222 are highly expressed in endothelial cellsReference Liu, Cheng, Yang, Xu and Zhang 231 and exert antiangiogenic, antiproliferative, antimigration and proapoptotic effects on endothelial cells,Reference Liu, Cheng, Yang, Xu and Zhang 231 which can be partly caused by reduced eNOS expression.Reference Suarez, Fernandez-Hernando, Pober and Sessa 232 In addition, miRNAs can modulate SIRT1 expression. Increased expression of miR-217 and miR-34a have been observed in endothelial senescence, which leads to loss of SIRT1 function, notably by reducing eNOS expression.Reference Xu, Seeger and Castillo 233

Conclusions

In individuals born after fetal growth restriction, early endothelial dysfunction plays an important role in the subsequent development of HTN, coronary heart disease and CKD. Decreased NO synthesis and bioavailability caused by defective eNOS function and oxidative stress, decreased EPCs number and function, and vascular senescence have all been shown to be involved in endothelial dysfunction (Fig. 1). Preventive approaches, including breastfeeding and supplementation with folate, vitamins, antioxidants, L-citrulline, L-Arg and NO modulators, represent promising and simple ways to prevent fetal growth restriction, improve endothelial function and vasodilation responses early in life and delay/prevent detrimental cardiovascular issues.

Fig. 1 Association between intrauterine growth restriction, early endothelial dysfunction and long-term cardiovascular-related diseases. eNOS, endothelial nitric oxide synthase; NO, nitric oxide synthase.

Epigenetic modulation of gene expression appears to be one of the main contributors to the long-term effects of an adverse perinatal environment. The identification of early biomarkers of endothelial dysfunction, especially epigenetic biomarkers, could allow early screening and follow-up of individuals at risk of developing CVD, thus contributing to the development of preventive and therapeutic strategies to avert the long-term effects of endothelial dysfunction in infants born after IUGR.

Acknowledgments

The authors thank Dolorès Mosig, Katya Nardou and Dr Fabienne Maurer for their invaluable contributions.

Financial Support

This research received no specific grant from any funding agency in the commercial or not-for-profit sectors.

Conflicts of interest

None.

Footnotes

These authors contributed equally to this work.

References

1. Brodsky, D, Christou, H. Current concepts in intrauterine growth restriction. J Intensive Care Med. 2004; 19, 307319.CrossRefGoogle ScholarPubMed
2. Jang, DG, Jo, YS, Lee, SJ, Kim, N, Lee, GS. Perinatal outcomes and maternal clinical characteristics in IUGR with absent or reversed end-diastolic flow velocity in the umbilical artery. Arch Gynecol Obstet. 2011; 284, 7378.CrossRefGoogle ScholarPubMed
3. Saleem, T, Sajjad, N, Fatima, S, et al. Intrauterine growth retardation – small events, big consequences. Ital J Pediatr. 2011; 37, 41.CrossRefGoogle ScholarPubMed
4. Mongelli, M, Gardosi, J. Symphysis-fundus height and pregnancy characteristics in ultrasound-dated pregnancies. Obstet Gynecol. 1999; 94, 591594.Google ScholarPubMed
5. Committee on Practice Bulletins--Gynecology ACoO, Gynecologists WDCUSA. Intrauterine growth restriction. Clinical management guidelines for obstetrician-gynecologists. American College of Obstetricians and Gynecologists. Int J Gynaecol Obstet. 2001; 72, 8596.Google Scholar
6. Alberry, M, Soothill, P. Management of fetal growth restriction. Arch Dis Child Fetal Neonatal Ed. 2007; 92, F62F67.CrossRefGoogle ScholarPubMed
7. Figueras, F, Gardosi, J. Intrauterine growth restriction: new concepts in antenatal surveillance, diagnosis, and management. Am J Obstet Gynecol. 2011; 204, 288300.CrossRefGoogle ScholarPubMed
8. Barker, ED, McAuliffe, FM, Alderdice, F, et al. The role of growth trajectories in classifying fetal growth restriction. Obstet Gynecol. 2013; 122, 248254.CrossRefGoogle ScholarPubMed
9. Villar, J, Cheikh Ismail, L, Victora, CG, et al. International standards for newborn weight, length, and head circumference by gestational age and sex: the Newborn Cross-Sectional Study of the INTERGROWTH-21st Project. Lancet. 2014; 384, 857868.CrossRefGoogle ScholarPubMed
10. Villar, J, Giuliani, F, Fenton, TR, et al. INTERGROWTH-21st very preterm size at birth reference charts. Lancet. 2016; 387, 844845.Google Scholar
11. Rosario, FJ, Jansson, N, Kanai, Y, et al. Maternal protein restriction in the rat inhibits placental insulin, mTOR, and STAT3 signaling and down-regulates placental amino acid transporters. Endocrinology. 2011; 152, 11191129.CrossRefGoogle ScholarPubMed
12. Johansson, M, Karlsson, L, Wennergren, M, Jansson, T, Powell, TL. Activity and protein expression of Na+/K+ ATPase are reduced in microvillous syncytiotrophoblast plasma membranes isolated from pregnancies complicated by intrauterine growth restriction. J Clin Endocrinol Metab. 2003; 88, 28312837.Google Scholar
13. Settle, P, Sibley, CP, Doughty, IM, et al. Placental lactate transporter activity and expression in intrauterine growth restriction. J Soc Gynecol Investig. 2006; 13, 357363.Google Scholar
14. Chaiworapongsa, T, Chaemsaithong, P, Yeo, L, Romero, R. Pre-eclampsia part 1: current understanding of its pathophysiology. Nat Rev Nephrol. 2014; 10, 466480.Google Scholar
15. Adams Waldorf, KM, McAdams, RM. Influence of infection during pregnancy on fetal development. Reproduction. 2013; 146, R151R162.Google Scholar
16. Derricott, H, Jones, RL, Heazell, AE. Investigating the association of villitis of unknown etiology with stillbirth and fetal growth restriction – a systematic review. Placenta. 2013; 34, 856862.Google Scholar
17. Baba, S, Wikstrom, AK, Stephansson, O, Cnattingius, S. Changes in snuff and smoking habits in Swedish pregnant women and risk for small for gestational age births. BJOG. 2013; 120, 456462.Google Scholar
18. Maruyama, H, Shinozuka, M, Kondoh, Y, et al. Thrombocytopenia in preterm infants with intrauterine growth restriction. Acta Med Okayama. 2008; 62, 313317.Google ScholarPubMed
19. Hall, JG. Review and hypothesis: syndromes with severe intrauterine growth restriction and very short stature – are they related to the epigenetic mechanism(s) of fetal survival involved in the developmental origins of adult health and disease? Am J Med Genet A. 2010; 152A, 512527.CrossRefGoogle Scholar
20. Malik, S, Cleves, MA, Zhao, W, et al. Association between congenital heart defects and small for gestational age. Pediatrics. 2007; 119, e976e982.CrossRefGoogle ScholarPubMed
21. Hillman, S, Peebles, DM, Williams, DJ. Paternal metabolic and cardiovascular risk factors for fetal growth restriction: a case-control study. Diabetes Care. 2013; 36, 16751680.Google Scholar
22. Li, J, Tsuprykov, O, Yang, X, Hocher, B. Paternal programming of offspring cardiometabolic diseases in later life. J Hypertens. 2016; 34, 21112126.Google Scholar
23. Minshall, RD, Tiruppathi, C, Vogel, SM, Malik, AB. Vesicle formation and trafficking in endothelial cells and regulation of endothelial barrier function. Histochem Cell Biol. 2002; 117, 105112.Google Scholar
24. Purnomowati, A, Kariadi, SH, Achmad, TH, Mose, JC, Setianto, B. Endothelial dysfunction in the young adult: a retrospective cohort study on the effect of low birth weight. Acta Med Indones. 2014; 46, 111116.Google Scholar
25. Bassareo, PP, Fanos, V, Puddu, M, et al. Reduced brachial flow-mediated vasodilation in young adult ex extremely low birth weight preterm: a condition predictive of increased cardiovascular risk? J Matern Fetal Neonatal Med. 2010; 23(Suppl. 3), 121124.Google Scholar
26. Martin, H, Lindblad, B, Norman, M. Endothelial function in newborn infants is related to folate levels and birth weight. Pediatrics. 2007; 119, 11521158.CrossRefGoogle ScholarPubMed
27. Leeson, P, Thorne, S, Donald, A, et al. Non-invasive measurement of endothelial function: effect on brachial artery dilatation of graded endothelial dependent and independent stimuli. Heart. 1997; 78, 2227.Google Scholar
28. Leeson, C, Whincup, P, Cook, D, et al. Flow-mediated dilation in 9- to 11-year-old children: the influence of intrauterine and childhood factors. Circulation. 1997; 96, 22332238.Google Scholar
29. Goodfellow, J, Bellamy, MF, Gorman, ST, et al. Endothelial function is impaired in fit young adults of low birth weight. Cardiovasc Res. 1998; 40, 600606.Google Scholar
30. Leeson, C, Kattenhorn, M, Morley, R, Lucas, A, Deanfield, J. Impact of low birth weight and cardiovascular risk factors on endothelial function in early adult life. Circulation. 2001; 103, 12641268.CrossRefGoogle ScholarPubMed
31. Krause, BJ, Carrasco-Wong, I, Caniuguir, A, et al. Endothelial eNOS/arginase imbalance contributes to vascular dysfunction in IUGR umbilical and placental vessels. Placenta. 2013; 34, 2028.CrossRefGoogle ScholarPubMed
32. Yzydorczyk, C, Gobeil, F Jr, Cambonie, G, et al. Exaggerated vasomotor response to ANG II in rats with fetal programming of hypertension associated with exposure to a low-protein diet during gestation. Am J Physiol Regul Integr Comp Physiol. 2006; 291, R1060R1068.CrossRefGoogle Scholar
33. Pladys, P, Sennlaub, F, Brault, S, et al. Microvascular rarefaction and decreased angiogenesis in rats with fetal programming of hypertension associated with exposure to a low-protein diet in utero. Am J Physiol Regul Integr Comp Physiol. 2005; 289, R1580R1588.CrossRefGoogle ScholarPubMed
34. Brawley, L, Itoh, S, Torrens, C, et al. Dietary protein restriction in pregnancy induces hypertension and vascular defects in rat male offspring. Pediatr Res. 2003; 54, 8390.Google Scholar
35. Franco, MC, Arruda, R, Dantas, A, et al. Intrauterine undernutrition: expression and activity of the endothelial nitric oxide synthase in male and female adult offspring. Cardiovasc Res. 2002; 56, 145153.Google Scholar
36. Tare, M, Parkington, HC, Wallace, EM, et al. Maternal melatonin administration mitigates coronary stiffness and endothelial dysfunction, and improves heart resilience to insult in growth restricted lambs. J Physiol. 2014; 592, 26952709.Google Scholar
37. Borwick, SC, Rhind, SM, McMillen, SR, Racey, PA. Effect of undernutrition of ewes from the time of mating on fetal ovarian development in mid gestation. Reprod Fertil Dev. 1997; 9, 711715.Google Scholar
38. Hurtado, R, Celani, M, Geber, S. Effect of short-term estrogen therapy on endothelial function: a double-blinded, randomized, controlled trial. Climacteric. 2016; 19, 448451.Google Scholar
39. Gleeson, M, Bishop, NC, Stensel, DJ, et al. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat Rev Immunol. 2011; 11, 607615.Google Scholar
40. Leinonen, E, Hurt-Camejo, E, Wiklund, O, et al. Insulin resistance and adiposity correlate with acute-phase reaction and soluble cell adhesion molecules in type 2 diabetes. Atherosclerosis. 2003; 166, 387394.Google Scholar
41. Pellanda, LC, Duncan, BB, Vigo, A, et al. Low birth weight and markers of inflammation and endothelial activation in adulthood: the ARIC study. Int J Cardiol. 2009; 134, 371377.Google Scholar
42. Teeninga, N, Schreuder, MF, Bokenkamp, A, Delemarre-van de Waal, HA, van Wijk, JA. Influence of low birth weight on minimal change nephrotic syndrome in children, including a meta-analysis. Nephrol Dial Transplant. 2008; 23, 16151620.Google Scholar
43. Skilton, MR, Evans, N, Griffiths, KA, Harmer, JA, Celermajer, DS. Aortic wall thickness in newborns with intrauterine growth restriction. Lancet. 2005; 365, 14841486.Google Scholar
44. Koklu, E, Ozturk, MA, Gunes, T, Akcakus, M, Kurtoglu, S. Is increased intima-media thickness associated with preatherosclerotic changes in intrauterine growth restricted newborns? Acta Paediatr. 2007; 96, 1858; author reply 1859.Google Scholar
45. Litwin, M, Niemirska, A. Intima-media thickness measurements in children with cardiovascular risk factors. Pediatr Nephrol. 2009; 24, 707719.Google Scholar
46. Cosmi, E, Visentin, S, Fanelli, T, Mautone, AJ, Zanardo, V. Aortic intima media thickness in fetuses and children with intrauterine growth restriction. Obstet Gynecol. 2009; 114, 11091114.Google Scholar
47. Crispi, F, Figueras, F, Cruz-Lemini, M, et al. Cardiovascular programming in children born small for gestational age and relationship with prenatal signs of severity. Am J Obstet Gynecol. 2012; 207, 121 e121121 e129.Google Scholar
48. Crispi, F, Bijnens, B, Figueras, F, et al. Fetal growth restriction results in remodeled and less efficient hearts in children. Circulation. 2010; 121, 24272436.CrossRefGoogle ScholarPubMed
49. Oren, A, Vos, LE, Uiterwaal, CS, et al. Birth weight and carotid intima-media thickness: new perspectives from the atherosclerosis risk in young adults (ARYA) study. Ann Epidemiol. 2004; 14, 816.Google Scholar
50. Jensen, GM, Moore, LG. The effect of high altitude and other risk factors on birthweight: independent or interactive effects? Am J Public Health. 1997; 87, 10031007.Google Scholar
51. Lueder, FL, Kim, SB, Buroker, CA, Bangalore, SA, Ogata, ES. Chronic maternal hypoxia retards fetal growth and increases glucose utilization of select fetal tissues in the rat. Metabolism. 1995; 44, 532537.CrossRefGoogle ScholarPubMed
52. Barker, DJ. The fetal origins of coronary heart disease. Acta Paediatr Suppl. 1997; 422, 7882.Google Scholar
53. Barker, DJ, Osmond, C, Golding, J, Kuh, D, Wadsworth, ME. Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ. 1989; 298, 564567.CrossRefGoogle ScholarPubMed
54. Giaccia, AJ, Simon, MC, Johnson, R. The biology of hypoxia: the role of oxygen sensing in development, normal function, and disease. Genes Dev. 2004; 18, 21832194.CrossRefGoogle ScholarPubMed
55. Malamitsi-Puchner, A, Boutsikou, T, Economou, E, et al. Angiopoietin-2 in the perinatal period and the role of intrauterine growth restriction. Acta Obstet Gynecol Scand. 2006; 85, 4548.Google Scholar
56. Griendling, KK, Harrison, DG. Dual role of reactive oxygen species in vascular growth. Circ Res. 1999; 85, 562563.CrossRefGoogle ScholarPubMed
57. Irani, K. Oxidant signaling in vascular cell growth, death, and survival: a review of the roles of reactive oxygen species in smooth muscle and endothelial cell mitogenic and apoptotic signaling. Circ Res. 2000; 87, 179183.Google Scholar
58. Touyz, RM, Schiffrin, EL. Reactive oxygen species in vascular biology: implications in hypertension. Histochem Cell Biol. 2004; 122, 339352.Google Scholar
59. Ushio-Fukai, M, Zafari, AM, Fukui, T, Ishizaka, N, Griendling, KK. p22phox is a critical component of the superoxide-generating NADH/NADPH oxidase system and regulates angiotensin II-induced hypertrophy in vascular smooth muscle cells. J Biol Chem. 1996; 271, 2331723321.Google Scholar
60. Griendling, KK, Minieri, CA, Ollerenshaw, JD, Alexander, RW. Angiotensin II stimulates NADH and NADPH oxidase activity in cultured vascular smooth muscle cells. Circ Res. 1994; 74, 11411148.Google Scholar
61. Zafari, AM, Ushio-Fukai, M, Akers, M, et al. Role of NADH/NADPH oxidase-derived H2O2 in angiotensin II-induced vascular hypertrophy. Hypertension. 1998; 32, 488495.Google Scholar
62. Tyagi, SC, Simon, SR. Regulation of neutrophil elastase activity by elastin-derived peptide. J Biol Chem. 1993; 268, 1651316518.Google Scholar
63. Chow, AK, Cena, J, Schulz, R. Acute actions and novel targets of matrix metalloproteinases in the heart and vasculature. Br J Pharmacol. 2007; 152, 189205.Google Scholar
64. Rajagopalan, S, Meng, XP, Ramasamy, S, Harrison, DG, Galis, ZS. Reactive oxygen species produced by macrophage-derived foam cells regulate the activity of vascular matrix metalloproteinases in vitro. Implications for atherosclerotic plaque stability. J Clin Invest. 1996; 98, 25722579.Google Scholar
65. Sesso, R, Franco, MC. Abnormalities in metalloproteinase pathways and IGF-I axis: a link between birth weight, hypertension, and vascular damage in childhood. Am J Hypertens. 2010; 23, 611.Google Scholar
66. Huyard, F, Yzydorczyk, C, Castro, MM, et al. Remodeling of aorta extracellular matrix as a result of transient high oxygen exposure in newborn rats: implication for arterial rigidity and hypertension risk. PLoS One. 2014; 9, e92287.Google Scholar
67. Yzydorczyk, C, Comte, B, Cambonie, G, et al. Neonatal oxygen exposure in rats leads to cardiovascular and renal alterations in adulthood. Hypertension. 2008; 52, 889895.Google Scholar
68. Mivelaz, Y YC, Barbier, A, Cloutier, A, Fouron, JC, Nuyt, AM. Neonatal oxygen exposure leads to increased aortic wall stiffness in adult rats: a Doppler ultrasound study. J Dev Orig Health Dis. 2011; 2, 184189.Google Scholar
69. Chatterjee, A, Black, SM, Catravas, JD. Endothelial nitric oxide (NO) and its pathophysiologic regulation. Vascul Pharmacol. 2008; 49, 134140.CrossRefGoogle ScholarPubMed
70. Förstermann, U, Münzel, T. Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation. 2006; 113, 17081714.Google Scholar
71. Searles, CD. Transcriptional and posttranscriptional regulation of endothelial nitric oxide synthase expression. Am J Physiol Cell Physiol. 2006; 291, C803C816.Google Scholar
72. De Caterina, R, Libby, P, Peng, HB, et al. Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J Clin Invest. 1995; 96, 6068.Google Scholar
73. Hata, T, Hashimoto, M, Manabe, A, et al. Maternal and fetal nitric oxide synthesis is decreased in pregnancies with small for gestational age infants. Hum Reprod. 1998; 13, 10701073.Google Scholar
74. Singh, S, Singh, A, Sharma, D, et al. Effect of L-arginine on nitric oxide levels in intrauterine growth restriction and its correlation with fetal outcome. Indian J Clin Biochem. 2015; 30, 298304.Google Scholar
75. Lyall, F, Greer, IA, Young, A, Myatt, L. Nitric oxide concentrations are increased in the feto-placental circulation in intrauterine growth restriction. Placenta. 1996; 17, 165168.Google Scholar
76. Myatt, L, Eis, AL, Brockman, DE, Greer, IA, Lyall, F. Endothelial nitric oxide synthase in placental villous tissue from normal, pre-eclamptic and intrauterine growth restricted pregnancies. Hum Reprod. 1997; 12, 167172.CrossRefGoogle ScholarPubMed
77. Payne, JA, Alexander, BT, Khalil, RA. Reduced endothelial vascular relaxation in growth-restricted offspring of pregnant rats with reduced uterine perfusion. Hypertension. 2003; 42, 768774.Google Scholar
78. Sathishkumar, K, Elkins, R, Yallampalli, U, Yallampalli, C. Protein restriction during pregnancy induces hypertension and impairs endothelium-dependent vascular function in adult female offspring. J Vasc Res. 2009; 46, 229239.Google Scholar
79. Bourdon, A, Parnet, P, Nowak, C, et al. l-Citrulline supplementation enhances fetal growth and protein synthesis in rats with intrauterine growth restriction. J Nutr. 2016; 146, 532541.Google Scholar
80. Sathishkumar, K, Elkins, R, Yallampalli, U, Balakrishnan, M, Yallampalli, C. Fetal programming of adult hypertension in female rat offspring exposed to androgens in utero. Early Hum Dev. 2011; 87, 407414.Google Scholar
81. Hracsko, Z, Hermesz, E, Ferencz, A, et al. Endothelial nitric oxide synthase is up-regulated in the umbilical cord in pregnancies complicated with intrauterine growth retardation. In Vivo. 2009; 23, 727732.Google Scholar
82. Dellee, U, Tobias, S, Li, H, Mildenberger, E. Expression of NO synthases and redox enzymes in umbilical arteries from newborns born small, appropriate, and large for gestational age. Pediatr Res. 2013; 73, 142146.Google Scholar
83. Takushima, S, Nishi, Y, Nonoshita, A, et al. Changes in the nitric oxide-soluble guanylate cyclase system and natriuretic peptide receptor system in placentas of pregnant Dahl salt-sensitive rats. J Obstet Gynaecol Res. 2015; 41, 540550.Google Scholar
84. Arroyo, JA, Anthony, RV, Parker, TA, Galan, HL. eNOS, NO, and the activation of ERK and AKT signaling at mid-gestation and near-term in an ovine model of intrauterine growth restriction. Syst Biol Reprod Med. 2010; 56, 6273.Google Scholar
85. Tolbert, T, Thompson, JA, Bouchard, P, Oparil, S. Estrogen-induced vasoprotection is independent of inducible nitric oxide synthase expression: evidence from the mouse carotid artery ligation model. Circulation. 2001; 104, 27402745.Google Scholar
86. Krause, BJ, Costello, PM, Munoz-Urrutia, E, et al. Role of DNA methyltransferase 1 on the altered eNOS expression in human umbilical endothelium from intrauterine growth restricted fetuses. Epigenetics. 2013; 8, 944952.CrossRefGoogle ScholarPubMed
87. Laskowska, M, Laskowska, K, Oleszczuk, J. Differences in the association between maternal serum homocysteine and ADMA levels in women with pregnancies complicated by preeclampsia and/or intrauterine growth restriction. Hypertens Pregnancy. 2013; 32, 8393.CrossRefGoogle ScholarPubMed
88. Gumus, E, Atalay, MA, Cetinkaya Demir, B, Sahin Gunes, E. Possible role of asymmetric dimethylarginine (ADMA) in prediction of perinatal outcome in preeclampsia and fetal growth retardation related to preeclampsia. J Matern Fetal Neonatal Med. 2016; 29, 38063811.Google Scholar
89. Rizos, D, Eleftheriades, M, Batakis, E, et al. Levels of asymmetric dimethylarginine throughout normal pregnancy and in pregnancies complicated with preeclampsia or had a small for gestational age baby. J Matern Fetal Neonatal Med. 2012; 25, 13111315.Google Scholar
90. Post, MS, Verhoeven, MO, van der Mooren, MJ, et al. Effect of hormone replacement therapy on plasma levels of the cardiovascular risk factor asymmetric dimethylarginine: a randomized, placebo-controlled 12-week study in healthy early postmenopausal women. J Clin Endocrinol Metab. 2003; 88, 42214226.CrossRefGoogle ScholarPubMed
91. Karkanaki, A, Vavilis, D, Traianos, A, Kalogiannidis, I, Panidis, D. Hormone therapy and asymmetrical dimethylarginine in postmenopausal women. Hormones (Athens). 2010; 9, 127135.Google Scholar
92. Yu, XJ, Li, YJ, Xiong, Y. Increase of an endogenous inhibitor of nitric oxide synthesis in serum of high cholesterol fed rabbits. Life Sci. 1994; 54, 753758.Google Scholar
93. Boger, RH, Bode-Boger, SM, Sydow, K, Heistad, DD, Lentz, SR. Plasma concentration of asymmetric dimethylarginine, an endogenous inhibitor of nitric oxide synthase, is elevated in monkeys with hyperhomocyst(e)inemia or hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2000; 20, 15571564.Google Scholar
94. Griendling, KK, FitzGerald, GA. Oxidative stress and cardiovascular injury: part I: basic mechanisms and in vivo monitoring of ROS. Circulation. 2003; 108, 19121916.Google Scholar
95. Burton, GJ, Jauniaux, E. Oxidative stress. Best Pract Res Clin Obstet Gynaecol. 2011; 25, 287299.Google Scholar
96. Takagi, Y, Nikaido, T, Toki, T, et al. Levels of oxidative stress and redox-related molecules in the placenta in preeclampsia and fetal growth restriction. Virchows Arch. 2004; 444, 4955.Google Scholar
97. Maisonneuve, E, Delvin, E, Edgard, A, et al. Oxidative conditions prevail in severe IUGR with vascular disease and Doppler anomalies. J Matern Fetal Neonatal Med. 2015; 28, 14711475.Google Scholar
98. Webster, RP, Roberts, VH, Myatt, L. Protein nitration in placenta – functional significance. Placenta. 2008; 29, 985994.Google Scholar
99. Kossenjans, W, Eis, A, Sahay, R, Brockman, D, Myatt, L. Role of peroxynitrite in altered fetal-placental vascular reactivity in diabetes or preeclampsia. Am J Physiol Heart Circ Physiol. 2000; 278, H1311H1319.Google Scholar
100. Santilli, F, D’Ardes, D, Davi, G. Oxidative stress in chronic vascular disease: from prediction to prevention. Vascul Pharmacol. 2015; 74, 2337.Google Scholar
101. Yzydorczyk, C, Comte, B, Huyard, F, et al. Developmental programming of eNOS uncoupling and enhanced vascular oxidative stress in adult rats after transient neonatal oxygen exposure. J Cardiovasc Pharmacol. 2013; 61, 816.Google Scholar
102. Vasquez-Vivar, J, Kalyanaraman, B, Martasek, P, et al. Superoxide generation by endothelial nitric oxide synthase: the influence of cofactors. Proc Natl Acad Sci U S A. 1998; 95, 92209225.Google Scholar
103. d’Uscio, LV, Santhanam, AV, Katusic, ZS. Erythropoietin prevents endothelial dysfunction in GTP-cyclohydrolase I-deficient hph1 mice. J Cardiovasc Pharmacol. 2014; 64, 514521.CrossRefGoogle ScholarPubMed
104. Yang, YM, Huang, A, Kaley, G, Sun, D. eNOS uncoupling and endothelial dysfunction in aged vessels. Am J Physiol Heart Circ Physiol. 2009; 297, H1829H1836.Google Scholar
105. Landmesser, U, Dikalov, S, Price, SR, et al. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest. 2003; 111, 12011209.Google Scholar
106. Chalupsky, K, Cai, H. Endothelial dihydrofolate reductase: critical for nitric oxide bioavailability and role in angiotensin II uncoupling of endothelial nitric oxide synthase. Proc Natl Acad Sci U S A. 2005; 102, 90569061.Google Scholar
107. Sydow, K, Munzel, T. ADMA and oxidative stress. Atheroscler Suppl. 2003; 4, 4151.Google Scholar
108. Schneider, D, Hernandez, C, Farias, M, et al. Oxidative stress as common trait of endothelial dysfunction in chorionic arteries from fetuses with IUGR and LGA. Placenta. 2015; 36, 552558.Google Scholar
109. Mitchell, BM, Cook, LG, Danchuk, S, Puschett, JB. Uncoupled endothelial nitric oxide synthase and oxidative stress in a rat model of pregnancy-induced hypertension. Am J Hypertens. 2007; 20, 12971304.Google Scholar
110. Oliveira, V, Akamine, EH, Carvalho, MH, et al. Influence of aerobic training on the reduced vasoconstriction to angiotensin II in rats exposed to intrauterine growth restriction: possible role of oxidative stress and AT2 receptor of angiotensin II. PLoS One. 2014; 9, e113035.Google Scholar
111. Asahara, T, Murohara, T, Sullivan, A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997; 275, 964967.Google Scholar
112. Yoder, MC, Mead, LE, Prater, D, et al. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood. 2007; 109, 18011809.Google Scholar
113. Purhonen, S, Palm, J, Rossi, D, et al. Bone marrow-derived circulating endothelial precursors do not contribute to vascular endothelium and are not needed for tumor growth. Proc Natl Acad Sci U S A. 2008; 105, 66206625.Google Scholar
114. Hill, JM, Zalos, G, Halcox, JP, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003; 348, 593600.Google Scholar
115. Sipos, PI, Crocker, IP, Hubel, CA, Baker, PN. Endothelial progenitor cells: their potential in the placental vasculature and related complications. Placenta. 2010; 31, 110.Google Scholar
116. Hwang, HS, Kwon, YG, Kwon, JY, et al. Senescence of fetal endothelial progenitor cell in pregnancy with idiopathic fetal growth restriction. J Matern Fetal Neonatal Med. 2012; 25, 17691773.Google Scholar
117. Ligi, I, Simoncini, S, Tellier, E, et al. A switch toward angiostatic gene expression impairs the angiogenic properties of endothelial progenitor cells in low birth weight preterm infants. Blood. 2011; 118, 16991709.Google Scholar
118. Ligi, I, Simoncini, S, Tellier, E, et al. Altered angiogenesis in low birth weight individuals: a role for anti-angiogenic circulating factors. J Matern Fetal Neonatal Med. 2014; 27, 233238.Google Scholar
119. Minamino, T, Komuro, I. Vascular cell senescence: contribution to atherosclerosis. Circ Res. 2007; 100, 1526.Google Scholar
120. Erusalimsky, JD, Fenton, M. Further in vivo evidence that cellular senescence is implicated in vascular pathophysiology. Circulation. 2002; 106, e144; author reply e144.Google Scholar
121. Borradaile, NM, Pickering, JG. NAD(+), sirtuins, and cardiovascular disease. Curr Pharm Des. 2009; 15, 110117.Google Scholar
122. Ota, H, Akishita, M, Eto, M, et al. Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol. 2007; 43, 571579.Google Scholar
123. Vassallo, PF, Simoncini, S, Ligi, I, et al. Accelerated senescence of cord blood endothelial progenitor cells in premature neonates is driven by SIRT1 decreased expression. Blood. 2014; 123, 21162126.Google Scholar
124. Fattal-Valevski, A, Bernheim, J, Leitner, Y, et al. Blood pressure values in children with intrauterine growth retardation. Isr Med Assoc J. 2001; 3, 805808.Google Scholar
125. Rossi, P, Tauzin, L, Marchand, E, et al. Respective roles of preterm birth and fetal growth restriction in blood pressure and arterial stiffness in adolescence. J Adolesc Health. 2011; 48, 520522.Google Scholar
126. Chiolero, A, Cachat, F, Burnier, M, Paccaud, F, Bovet, P. Prevalence of hypertension in schoolchildren based on repeated measurements and association with overweight. J Hypertens. 2007; 25, 22092217.Google Scholar
127. Leon, DA, Johansson, M, Rasmussen, F. Gestational age and growth rate of fetal mass are inversely associated with systolic blood pressure in young adults: an epidemiologic study of 165,136 Swedish men aged 18 years. Am J Epidemiol. 2000; 152, 597604.CrossRefGoogle ScholarPubMed
128. Nilsson, PM, Ostergren, PO, Nyberg, P, Soderstrom, M, Allebeck, P. Low birth weight is associated with elevated systolic blood pressure in adolescence: a prospective study of a birth cohort of 149378 Swedish boys. J Hypertens. 1997; 15, 16271631.Google Scholar
129. Gennser, G, Rymark, P, Isberg, PE. Low birth weight and risk of high blood pressure in adulthood. Br Med J (Clin Res Ed). 1988; 296, 14981500.Google Scholar
130. Martyn, CN, Barker, DJ, Jespersen, S, et al. Growth in utero, adult blood pressure, and arterial compliance. Br Heart J. 1995; 73, 116121.Google Scholar
131. Curhan, GC, Willett, WC, Rimm, EB, et al. Birth weight and adult hypertension, diabetes mellitus, and obesity in US men. Circulation. 1996; 94, 32463250.Google Scholar
132. Law, CM, Shiell, AW. Is blood pressure inversely related to birth weight? The strength of evidence from a systematic review of the literature. J Hypertens. 1996; 14, 935941.Google Scholar
133. Huxley, R, Neil, A, Collins, R. Unravelling the fetal origins hypothesis: is there really an inverse association between birthweight and subsequent blood pressure? Lancet. 2002; 360, 659665.Google Scholar
134. Tauzin, L, Rossi, P, Grosse, C, et al. Increased systemic blood pressure and arterial stiffness in young adults born prematurely. J Dev Orig Health Dis. 2014; 5, 448452.Google Scholar
135. Wlodek, ME, Westcott, K, Siebel, AL, Owens, JA, Moritz, KM. Growth restriction before or after birth reduces nephron number and increases blood pressure in male rats. Kidney Int. 2008; 74, 187195.Google Scholar
136. Alexander, BT. Placental insufficiency leads to development of hypertension in growth-restricted offspring. Hypertension. 2003; 41, 457462.Google Scholar
137. Bourque, SL, Gragasin, FS, Quon, AL, et al. Prenatal hypoxia causes long-term alterations in vascular endothelin-1 function in aged male, but not female, offspring. Hypertension. 2013; 62, 753758.Google Scholar
138. Ortiz, LA, Quan, A, Zarzar, F, Weinberg, A, Baum, M. Prenatal dexamethasone programs hypertension and renal injury in the rat. Hypertension. 2003; 41, 328334.Google Scholar
139. Mossa, F, Carter, F, Walsh, SW, et al. Maternal undernutrition in cows impairs ovarian and cardiovascular systems in their offspring. Biol Reprod. 2013; 88, 92.Google Scholar
140. Goyal, R, Van-Wickle, J, Goyal, D, Longo, LD. Antenatal maternal low protein diet: ACE-2 in the mouse lung and sexually dimorphic programming of hypertension. BMC Physiol. 2015; 15, 2.CrossRefGoogle ScholarPubMed
141. Gilbert, JS, Lang, AL, Grant, AR, Nijland, MJ. Maternal nutrient restriction in sheep: hypertension and decreased nephron number in offspring at 9 months of age. J Physiol. 2005; 565, 137147.Google Scholar
142. Ozaki, T, Nishina, H, Hanson, M, Poston, L. Dietary restriction in pregnant rats causes gender-related hypertension and vascular dysfunction in offspring. J Physiol. 2001; 530, 141152.Google Scholar
143. Cambonie, G, Comte, B, Yzydorczyk, C, et al. Antenatal antioxidant prevents adult hypertension, vascular dysfunction, and microvascular rarefaction associated with in utero exposure to a low-protein diet. Am J Physiol Regul Integr Comp Physiol. 2007; 292, R1236R1245.Google Scholar
144. Taddei, S, Virdis, A, Mattei, P, Arzilli, F, Salvetti, A. Endothelium-dependent forearm vasodilation is reduced in normotensive subjects with familial history of hypertension. J Cardiovasc Pharmacol. 1992; 20(Suppl. 12), S193S195.Google Scholar
145. Miller, MJ, Pinto, A, Mullane, KM. Impaired endothelium-dependent relaxations in rabbits subjected to aortic coarctation hypertension. Hypertension. 1987; 10, 164170.Google Scholar
146. d’Uscio, LV, Barton, M, Shaw, S, Moreau, P, Luscher, TF. Structure and function of small arteries in salt-induced hypertension: effects of chronic endothelin-subtype-A-receptor blockade. Hypertension. 1997; 30, 905911.CrossRefGoogle ScholarPubMed
147. Verma, S, Anderson, TJ. Fundamentals of endothelial function for the clinical cardiologist. Circulation. 2002; 105, 546549.Google Scholar
148. Ludmer, PL, Selwyn, AP, Shook, TL, et al. Paradoxical vasoconstriction induced by acetylcholine in atherosclerotic coronary arteries. N Engl J Med. 1986; 315, 10461051.Google Scholar
149. Barker, DJ, Gluckman, PD, Godfrey, KM, et al. Fetal nutrition and cardiovascular disease in adult life. Lancet. 1993; 341, 938941.Google Scholar
150. Leon, DA, Lithell, HO, Vagero, D, et al. Reduced fetal growth rate and increased risk of death from ischaemic heart disease: cohort study of 15 000 Swedish men and women born 1915-29. BMJ. 1998; 317, 241245.Google Scholar
151. Wang, SF, Shu, L, Sheng, J, et al. Birth weight and risk of coronary heart disease in adults: a meta-analysis of prospective cohort studies. J Dev Orig Health Dis. 2014; 5, 408419.Google Scholar
152. Eriksson, M, Tibblin, G, Cnattingius, S. Low birthweight and ischaemic heart disease. Lancet. 1994; 343, 731.Google Scholar
153. Banci, M, Saccucci, P, Dofcaci, A, et al. Birth weight and coronary artery disease. The effect of gender and diabetes. Int J Biol Sci. 2009; 5, 244248.Google Scholar
154. Abrahamson, DR, Robert, B, Hyink, DP, St John, PL, Daniel, TO. Origins and formation of microvasculature in the developing kidney. Kidney Int Suppl. 1998; 67, S7S11.Google Scholar
155. Hyink, DP, Tucker, DC, St John, PL, et al. Endogenous origin of glomerular endothelial and mesangial cells in grafts of embryonic kidneys. Am J Physiol. 1996; 270, F886F899.Google Scholar
156. Stehouwer, CD, Henry, RM, Dekker, JM, et al. Microalbuminuria is associated with impaired brachial artery, flow-mediated vasodilation in elderly individuals without and with diabetes: further evidence for a link between microalbuminuria and endothelial dysfunction – the Hoorn Study. Kidney Int Suppl. 2004; 66, S42S44.Google Scholar
157. Pedrinelli, R, Giampietro, O, Carmassi, F, et al. Microalbuminuria and endothelial dysfunction in essential hypertension. Lancet. 1994; 344, 1418.Google Scholar
158. Mancuso, P, Antoniotti, P, Quarna, J, et al. Validation of a standardized method for enumerating circulating endothelial cells and progenitors: flow cytometry and molecular and ultrastructural analyses. Clin Cancer Res. 2009; 15, 267273.Google Scholar
159. Perticone, F, Maio, R, Perticone, M, et al. Endothelial dysfunction and subsequent decline in glomerular filtration rate in hypertensive patients. Circulation. 2010; 122, 379384.Google Scholar
160. Gris, JC, Branger, B, Vecina, F, et al. Increased cardiovascular risk factors and features of endothelial activation and dysfunction in dialyzed uremic patients. Kidney Int. 1994; 46, 807813.Google Scholar
161. Manalich, R, Reyes, L, Herrera, M, Melendi, C, Fundora, I. Relationship between weight at birth and the number and size of renal glomeruli in humans: a histomorphometric study. Kidney Int. 2000; 58, 770773.Google Scholar
162. White, SL, Perkovic, V, Cass, A, et al. Is low birth weight an antecedent of CKD in later life? A systematic review of observational studies. Am J Kidney Dis. 2009; 54, 248261.Google Scholar
163. Giapros, V, Papadimitriou, P, Challa, A, Andronikou, S. The effect of intrauterine growth retardation on renal function in the first two months of life. Nephrol Dial Transplant. 2007; 22, 96103.Google Scholar
164. Silverwood, RJ, Pierce, M, Hardy, R, et al. Low birth weight, later renal function, and the roles of adulthood blood pressure, diabetes, and obesity in a British birth cohort. Kidney Int. 2013; 84, 12621270.Google Scholar
165. Vehaskari, VM, Aviles, DH, Manning, J. Prenatal programming of adult hypertension in the rat. Kidney Int. 2001; 59, 238245.Google Scholar
166. Woods, LL, Ingelfinger, JR, Nyengaard, JR, Rasch, R. Maternal protein restriction suppresses the newborn renin-angiotensin system and programs adult hypertension in rats. Pediatr Res. 2001; 49, 460467.Google Scholar
167. Boubred, F, Delamaire, E, Buffat, C, et al. High protein intake in neonatal period induces glomerular hypertrophy and sclerosis in adulthood in rats born with IUGR. Pediatr Res. 2016; 79, 2226.Google Scholar
168. Boubred, F, Buffat, C, Feuerstein, JM, et al. Effects of early postnatal hypernutrition on nephron number and long-term renal function and structure in rats. Am J Physiol Renal Physiol. 2007; 293, F1944F1949.Google Scholar
169. Boubred, F, Daniel, L, Buffat, C, et al. Early postnatal overfeeding induces early chronic renal dysfunction in adult male rats. Am J Physiol Renal Physiol. 2009; 297, F943F951.Google Scholar
170. Anderson, S, King, AJ, Brenner, BM. Hyperlipidemia and glomerular sclerosis: an alternative viewpoint. Am J Med. 1989; 87, 34N38N.Google Scholar
171. Ikeda, Y, Tajima, S, Izawa-Ishizawa, Y, et al. Bovine milk-derived lactoferrin exerts proangiogenic effects in an Src-Akt-eNOS-dependent manner in response to ischemia. J Cardiovasc Pharmacol. 2013; 61, 423429.Google Scholar
172. Safaeian, L, Javanmard, SH, Mollanoori, Y, Dana, N. Cytoprotective and antioxidant effects of human lactoferrin against H2O2-induced oxidative stress in human umbilical vein endothelial cells. Adv Biomed Res. 2015; 4, 188.Google Scholar
173. Verhaar, MC, Stroes, E, Rabelink, TJ. Folates and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2002; 22, 613.Google Scholar
174. Robinson, K, Arheart, K, Refsum, H, et al. Low circulating folate and vitamin B6 concentrations: risk factors for stroke, peripheral vascular disease, and coronary artery disease. European COMAC Group. Circulation. 1998; 97, 437443.Google Scholar
175. Antoniades, C, Shirodaria, C, Warrick, N, et al. 5-methyltetrahydrofolate rapidly improves endothelial function and decreases superoxide production in human vessels: effects on vascular tetrahydrobiopterin availability and endothelial nitric oxide synthase coupling. Circulation. 2006; 114, 11931201.Google Scholar
176. Xia, XS, Li, X, Wang, L, et al. Supplementation of folic acid and vitamin B(1)(2) reduces plasma levels of asymmetric dimethylarginine in patients with acute ischemic stroke. J Clin Neurosci. 2014; 21, 15861590.Google Scholar
177. Wu, CJ, Wang, L, Li, X, et al. Impact of adding folic acid, vitamin B(12) and probucol to standard antihypertensive medication on plasma homocysteine and asymmetric dimethylarginine levels of essential hypertension patients. Zhonghua Xin Xue Guan Bing Za Zhi. 2012; 40, 10031008.Google Scholar
178. Li, JM, Qu, PF, Dang, SN, et al. Effect of folic acid supplementation in childbearing aged women during pregnancy on neonate birth weight in Shaanxi province. Zhonghua Liu Xing Bing Xue Za Zhi. 2016; 37, 10171020.Google Scholar
179. Alessio, AC, Santos, CX, Debbas, V, et al. Evaluation of mild hyperhomocysteinemia during the development of atherosclerosis in apolipoprotein E-deficient and normal mice. Exp Mol Pathol. 2011; 90, 4550.Google Scholar
180. Torrens, C, Brawley, L, Anthony, FW, et al. Folate supplementation during pregnancy improves offspring cardiovascular dysfunction induced by protein restriction. Hypertension. 2006; 47, 982987.Google Scholar
181. Stroes, ES, van Faassen, EE, Yo, M, et al. Folic acid reverts dysfunction of endothelial nitric oxide synthase. Circ Res. 2000; 86, 11291134.Google Scholar
182. Zingg, JM, Azzi, A. Non-antioxidant activities of vitamin E. Curr Med Chem. 2004; 11, 11131133.Google Scholar
183. Wu, D, Liu, L, Meydani, M, Meydani, SN. Vitamin E increases production of vasodilator prostanoids in human aortic endothelial cells through opposing effects on cyclooxygenase-2 and phospholipase A2. J Nutr. 2005; 135, 18471853.Google Scholar
184. Tran, K, Chan, AC. R,R,R-alpha-tocopherol potentiates prostacyclin release in human endothelial cells. Evidence for structural specificity of the tocopherol molecule. Biochim Biophys Acta. 1990; 1043, 189197.Google Scholar
185. Memon, S, Pratten, MK. Developmental toxicity of ethanol in chick heart in ovo and in micromass culture can be prevented by addition of vitamin C and folic acid. Reprod Toxicol. 2009; 28, 262269.Google Scholar
186. Hovdenak, N, Haram, K. Influence of mineral and vitamin supplements on pregnancy outcome. Eur J Obstet Gynecol Reprod Biol. 2012; 164, 127132.Google Scholar
187. Sesso, HD, Buring, JE, Christen, WG, et al. Vitamins E and C in the prevention of cardiovascular disease in men: the Physicians’ Health Study II randomized controlled trial. JAMA. 2008; 300, 21232133.Google Scholar
188. Rumbold, A, Ota, E, Nagata, C, Shahrook, S, Crowther, CA. Vitamin C supplementation in pregnancy. Cochrane Database Syst Rev. 2015; 29, CD004072.Google Scholar
189. Rumbold, AR, Crowther, CA, Haslam, RR, et al. Vitamins C and E and the risks of preeclampsia and perinatal complications. N Engl J Med. 2006; 354, 17961806.Google Scholar
190. Care, AS, Sung, MM, Panahi, S, et al. Perinatal resveratrol supplementation to spontaneously hypertensive rat dams mitigates the development of hypertension in adult offspring. Hypertension. 2016; 67, 10381044.Google Scholar
191. Vaziri, ND, Ding, Y, Ni, Z, Gonick, HC. Altered nitric oxide metabolism and increased oxygen free radical activity in lead-induced hypertension: effect of lazaroid therapy. Kidney Int. 1997; 52, 10421046.Google Scholar
192. Vaziri, ND, Ni, Z, Oveisi, F, Trnavsky-Hobbs, DL. Effect of antioxidant therapy on blood pressure and NO synthase expression in hypertensive rats. Hypertension. 2000; 36, 957964.Google Scholar
193. Herrera, EA, Cifuentes-Zuniga, F, Figueroa, E, et al. N-acetylcysteine, a glutathione precursor, reverts vascular dysfunction and endothelial epigenetic programming in intrauterine growth restricted guinea pigs. J Physiol. 2017, 595, 10771092.Google Scholar
194. Hardeland, R, Cardinali, DP, Srinivasan, V, et al. Melatonin – a pleiotropic, orchestrating regulator molecule. Prog Neurobiol. 2011; 93, 350384.Google Scholar
195. Reiter, RJ, Tan, DX, Terron, MP, Flores, LJ, Czarnocki, Z. Melatonin and its metabolites: new findings regarding their production and their radical scavenging actions. Acta Biochim Pol. 2007; 54, 19.Google Scholar
196. Franco Mdo, C, Akamine, EH, Aparecida de Oliveira, M, et al. Vitamins C and E improve endothelial dysfunction in intrauterine-undernourished rats by decreasing vascular superoxide anion concentration. J Cardiovasc Pharmacol. 2003; 42, 211217.Google Scholar
197. Galano, A, Tan, DX, Reiter, RJ. On the free radical scavenging activities of melatonin’s metabolites, AFMK and AMK. J Pineal Res. 2013; 54, 245257.Google Scholar
198. Lopez, A, Garcia, JA, Escames, G, et al. Melatonin protects the mitochondria from oxidative damage reducing oxygen consumption, membrane potential, and superoxide anion production. J Pineal Res. 2009; 46, 188198.Google Scholar
199. Herrera, EA, Macchiavello, R, Montt, C, et al. Melatonin improves cerebrovascular function and decreases oxidative stress in chronically hypoxic lambs. J Pineal Res. 2014; 57, 3342.Google Scholar
200. Weekley, LB. Effects of melatonin on isolated pulmonary artery and vein: role of the vascular endothelium. Pulm Pharmacol. 1993; 6, 149154.Google Scholar
201. Girouard, H, Chulak, C, Lejossec, M, Lamontagne, D, de Champlain, J. Vasorelaxant effects of the chronic treatment with melatonin on mesenteric artery and aorta of spontaneously hypertensive rats. J Hypertens. 2001; 19, 13691377.Google Scholar
202. Das, R, Balonan, L, Ballard, HJ, Ho, S. Chronic hypoxia inhibits the antihypertensive effect of melatonin on pulmonary artery. Int J Cardiol. 2008; 126, 340345.Google Scholar
203. Curis, E, Nicolis, I, Moinard, C, et al. Almost all about citrulline in mammals. Amino Acids. 2005; 29, 177205.Google Scholar
204. Romero, MJ, Platt, DH, Caldwell, RB, Caldwell, RW. Therapeutic use of citrulline in cardiovascular disease. Cardiovasc Drug Rev. 2006; 24, 275290.Google Scholar
205. Chien, SJ, Lin, KM, Kuo, HC, et al. Two different approaches to restore renal nitric oxide and prevent hypertension in young spontaneously hypertensive rats: l-citrulline and nitrate. Transl Res. 2014; 163, 4352.Google Scholar
206. Figueroa, A, Trivino, JA, Sanchez-Gonzalez, MA, Vicil, F. Oral L-citrulline supplementation attenuates blood pressure response to cold pressor test in young men. Am J Hypertens. 2010; 23, 1216.Google Scholar
207. Xuan, C, Lun, LM, Zhao, JX, et al. L-citrulline for protection of endothelial function from ADMA-induced injury in porcine coronary artery. Sci Rep. 2015; 5, 10987.Google Scholar
208. Chen, J, Gong, X, Chen, P, Luo, K, Zhang, X. Effect of L-arginine and sildenafil citrate on intrauterine growth restriction fetuses: a meta-analysis. BMC Pregnancy Childbirth. 2016; 16, 225.Google Scholar
209. Gui, S, Jia, J, Niu, X, et al. Arginine supplementation for improving maternal and neonatal outcomes in hypertensive disorder of pregnancy: a systematic review. J Renin Angiotensin Aldosterone Syst. 2014; 15, 8896.Google Scholar
210. Vadillo-Ortega, F, Perichart-Perera, O, Espino, S, et al. Effect of supplementation during pregnancy with L-arginine and antioxidant vitamins in medical food on pre-eclampsia in high risk population: randomised controlled trial. BMJ. 2011; 342, d2901.CrossRefGoogle ScholarPubMed
211. Wu, G, Bazer, FW, Cudd, TA, et al. Pharmacokinetics and safety of arginine supplementation in animals. J Nutr. 2007; 137, 1673S1680S.Google Scholar
212. Dastjerdi, MV, Hosseini, S, Bayani, L. Sildenafil citrate and uteroplacental perfusion in fetal growth restriction. J Res Med Sci. 2012; 17, 632636.Google Scholar
213. Wareing, M, Myers, JE, O’Hara, M, Baker, PN. Sildenafil citrate (Viagra) enhances vasodilatation in fetal growth restriction. J Clin Endocrinol Metab. 2005; 90, 25502555.Google Scholar
214. Lassala, A, Bazer, FW, Cudd, TA, et al. Parenteral administration of L-arginine prevents fetal growth restriction in undernourished ewes. J Nutr. 2010; 140, 12421248.Google Scholar
215. Herraiz, S, Pellicer, B, Serra, V, et al. Sildenafil citrate improves perinatal outcome in fetuses from pre-eclamptic rats. BJOG. 2012; 119, 13941402.Google Scholar
216. Refuerzo, JS, Sokol, RJ, Aranda, JV, et al. Sildenafil citrate and fetal outcome in pregnant rats. Fetal Diagn Ther. 2006; 21, 259263.Google Scholar
217. Cutfield, WS, Hofman, PL, Mitchell, M, Morison, IM. Could epigenetics play a role in the developmental origins of health and disease? Pediatr Res. 2007; 61, 68R75R.Google Scholar
218. Chen, M, Zhang, L. Epigenetic mechanisms in developmental programming of adult disease. Drug Discov Today. 2011; 16, 10071018.Google Scholar
219. McKay, JA, Mathers, JC. Diet induced epigenetic changes and their implications for health. Acta Physiol (Oxf). 2011; 202, 103118.Google Scholar
220. Kangaspeska, S, Stride, B, Metivier, R, et al. Transient cyclical methylation of promoter DNA. Nature. 2008; 452, 112115.Google Scholar
221. Lorenzen, JM, Martino, F, Thum, T. Epigenetic modifications in cardiovascular disease. Basic Res Cardiol. 2012; 107, 245.Google Scholar
222. Ito, S, D’Alessio, AC, Taranova, OV, et al. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010; 466, 11291133.Google Scholar
223. Kriaucionis, S, Heintz, N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science. 2009; 324, 929930.Google Scholar
224. Postberg, J, Kanders, M, Forcob, S, et al. CpG signalling, H2A.Z/H3 acetylation and microRNA-mediated deferred self-attenuation orchestrate foetal NOS3 expression. Clin Epigenetics. 2015; 7, 9.Google Scholar
225. Canani, RB, Costanzo, MD, Leone, L, et al. Epigenetic mechanisms elicited by nutrition in early life. Nutr Res Rev. 2011; 24, 198205.Google Scholar
226. Xu, XF, Xu, SS, Fu, LC, et al. Epigenetic changes in peripheral leucocytes as biomarkers in intrauterine growth retardation rat. Biomed Rep. 2016; 5, 548552.Google Scholar
227. Shruti, K, Shrey, K, Vibha, R. Micro RNAs: tiny sequences with enormous potential. Biochem Biophys Res Commun. 2011; 407, 445449.Google Scholar
228. Sayed, D, Abdellatif, M. MicroRNAs in development and disease. Physiol Rev. 2011; 91, 827887.Google Scholar
229. Weber, M, Baker, MB, Moore, JP, Searles, CD. MiR-21 is induced in endothelial cells by shear stress and modulates apoptosis and eNOS activity. Biochem Biophys Res Commun. 2010; 393, 643648.Google Scholar
230. Fleissner, F, Jazbutyte, V, Fiedler, J, et al. Short communication: asymmetric dimethylarginine impairs angiogenic progenitor cell function in patients with coronary artery disease through a microRNA-21-dependent mechanism. Circ Res. 2010; 107, 138143.Google Scholar
231. Liu, X, Cheng, Y, Yang, J, Xu, L, Zhang, C. Cell-specific effects of miR-221/222 in vessels: molecular mechanism and therapeutic application. J Mol Cell Cardiol. 2012; 52, 245255.Google Scholar
232. Suarez, Y, Fernandez-Hernando, C, Pober, JS, Sessa, WC. Dicer dependent microRNAs regulate gene expression and functions in human endothelial cells. Circ Res. 2007; 100, 11641173.Google Scholar
233. Xu, Q, Seeger, FH, Castillo, J, et al. Micro-RNA-34a contributes to the impaired function of bone marrow-derived mononuclear cells from patients with cardiovascular disease. J Am Coll Cardiol. 2012; 59, 21072117.Google Scholar
Figure 0

Fig. 1 Association between intrauterine growth restriction, early endothelial dysfunction and long-term cardiovascular-related diseases. eNOS, endothelial nitric oxide synthase; NO, nitric oxide synthase.