Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Pediatr. May 8, 2016; 5(2): 172-181
Published online May 8, 2016. doi: 10.5409/wjcp.v5.i2.172
Fetal programming and early identification of newborns at high risk of free radical-mediated diseases
Serafina Perrone, Antonino Santacroce, Anna Picardi, Giuseppe Buonocore, Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
Author contributions: All authors equally contributed to this paper for conception, design of the study, literature review, analysis, drafting, critical revision, editing, and final approval of the final version.
Conflict-of-interest statement: The authors confirm that this article content has no conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Serafina Perrone, MD, PhD, Department of Molecular and Developmental Medicine, University of Siena, Policlinico Santa Maria alle Scotte, Viale Bracci 36, 53100 Siena, Italy. saraspv@yahoo.it
Telephone: +39-0577-586542 Fax: +39-0577-586182
Received: August 29, 2015
Peer-review started: September 6, 2015
First decision: October 8, 2015
Revised: January 25, 2016
Accepted: February 14, 2016
Article in press: February 16, 2016
Published online: May 8, 2016

Abstract

Nowadays metabolic syndrome represents a real outbreak affecting society. Paradoxically, pediatricians must feel involved in fighting this condition because of the latest evidences of developmental origins of adult diseases. Fetal programming occurs when the normal fetal development is disrupted by an abnormal insult applied to a critical point in intrauterine life. Placenta assumes a pivotal role in programming the fetal experience in utero due to the adaptive changes in structure and function. Pregnancy complications such as diabetes, intrauterine growth restriction, pre-eclampsia, and hypoxia are associated with placental dysfunction and programming. Many experimental studies have been conducted to explain the phenotypic consequences of fetal-placental perturbations that predispose to the genesis of metabolic syndrome, obesity, diabetes, hyperinsulinemia, hypertension, and cardiovascular disease in adulthood. In recent years, elucidating the mechanisms involved in such kind of process has become the challenge of scientific research. Oxidative stress may be the general underlying mechanism that links altered placental function to fetal programming. Maternal diabetes, prenatal hypoxic/ischaemic events, inflammatory/infective insults are specific triggers for an acute increase in free radicals generation. Early identification of fetuses and newborns at high risk of oxidative damage may be crucial to decrease infant and adult morbidity.

Key Words: Fetal programming, Oxidative stress, High-risk newborn, Biomarkers, Perinatal medicine, Metabolic syndrome

Core tip: The adverse outcomes on the offspring born from altered gestation are already known. The consequences of these perturbations have been demonstrated even after many decades from birth. In this review we summarize gestational conditions associated to fetal programming and elucidate the mechanisms involved in such kind of occurrence. We also describe to what extent oxidative stress (OS) is involved in a very wide spectrum of genetic, metabolic, and cellular responses, through the gene expression regulation, and cell growth modulation. By virtue of these properties, OS has been nominated as the lowest common denominator of adult disease programming.



INTRODUCTION

The last century witnessed the rise in chronic cardio-metabolic diseases in which metabolic-syndrome (MetS) represents a major health problem regarding morbidity and mortality[1]. MetS is characterized by a number of related disorders, such as visceral obesity, glucose intolerance, disturbed plasma lipids concentration, high blood pressure, and increased risk of developing cardiovascular diseases and type 2 diabetes[2]. Smoking, high-fat diets, abdominal obesity[3-5], insulin resistance[6,7], physical inactivity[4,8], aging[9], and hormonal imbalance[10] have been identified as the main risk factors for several years.

Pediatricians have serious concerns with MetS because adult lifestyle is not the only determinant. In the last decades, a worldwide series of epidemiological studies have provided evidence for the association between perturbation of fetal environment and major risk factors for cardiovascular disease, diabetes, and MetS in adult life[11-15]. This has been called “fetal/early origins of adult disease” by David Barker. The hypothesis predicts that environmental factors, particularly nutrition, act in early life to program the risks for adverse health outcomes later in life[16]. Refinements of this idea of “fetal programming” focus on the processes of developmental plasticity, which in normal situations provide the settings for homeostatic mechanisms to ensure an adequate amount of nutrients to the most vital organs at the expenses of other less vital organs (the thrifty phenotype hypothesis)[17]. These changes in phenotype can become permanent and can generate a mismatch with adult environment that would lead to the development of metabolic diseases in adulthood[18]. The latter phenomenon gave rise to the new concepts of “metabolic memory”[19], “fetal primed”[20], and “developmental plasticity”[21].

The aim of this paper is to review all the gestational conditions associated to fetal programming and elucidate mechanisms involved in such kind of process. Identifying a lowest common denominator could be essential to contrive prevention strategies, treatment, and appropriate follow-up to high-risk newborns.

FETAL PROGRAMMING

Fetal programming occurs when the normal pattern of fetal development is disrupted by an abnormal stimulus or insult applied to a critical point in intrauterine life. Pregnancies complicated by diabetes, small for gestational age (SGA) or large for gestational age (LGA) offspring, pre-eclampsia and conditions such as hypoxia, oxidative and nitrosative stress are associated with programming. Placenta plays a key role in developmental plasticity. Vasculature and trophoblast are both involved in overall placental transport[22,23]. Changing developmental signals or the amount of substrate of the fetus produces an alteration of fetal development which ultimately leads to cardiovascular or metabolic diseases later in adult life[24]. Alterations in placental vasculogenesis[25], trophoblast expression of transporters[26], trophoblast enzyme activity, and hormone production[27] occur in pregnancies complicated by IUGR, pre-eclampsia or diabetes.

Mothers with insulin-dependent diabetes are prone to hyperglycemia in the first trimester of gestation that generates an up-regulation of Glut1 and System A (a sodium-dependent transporter of neutral amino acid) in the trophoblast leading to accelerated fetal growth in late gestation[28]. The activity of System A is reduced in placentas with intrauterine growth restriction (IUGR)[29,30]; moreover, inhibition of System A in rats causes growth restriction[31]. Glut transporters function and expression are also influenced by glucocorticoids, which are produced by trophoblast and regulated by the activity of 11-β-hydroxysteroid dehydrogenase (11βHSD). Exposure of the rat fetus to excess maternal or exogenous glucocorticoids causes growth restriction, hypertension and hyperglycaemia[32,33]. The trophoblast expresses 11βHSD-2 that converts cortisol to inactive cortisone and this may protect the fetus against high levels of maternal cortisol[34]. In humans, mutations in the 11βHSD-2 gene have been reported in association with low birth weight. Reduced 11βHSD-2 activity and increased fetal cortisol levels have been reported in association with IUGR[35].

Hypoxic conditions in pregnancy are strongly involved in fetal programming. Oxygen regulates development of the villous vascular tree and villous trophoblast proliferation due to hypoxic regulation of angiogenic mediators as vascular endothelial growth factor (VEGF) and placental growth factor (PLGF). Hypoxia acts via the transcription of hypoxia-inducible factor-1α (HIF-1α) that activates gene transcription in response to varying oxygen concentration. For example, at 10-12 wk of gestation, the trophoblast is exposed to a hyperoxic challenge during the transition from histiotrophic nutrition to intervillous blood flow vascularization[36]. Low oxygen tension inhibits trophoblast differentiation to the invasive extravillous trophoblast pathway, hence the switch in oxygenation activates trophoblast invasion and subjects the cell to oxidative and nitrosative stress. A pathological increase of oxidative stress (OS) is found in pregnancy complicated by pre-eclampsia or diabetes[37].

On the basis of the latter consideration, in order to confirm the hypothesis of in utero programming process and analyze the mechanisms involved, many authors have conducted experimental studies throughout various animal models of fetal programming based on fetal insult induced by placental insufficiency, hypoxia, maternal undernutrition, and maternal exposure to stress and increased plasma glucocorticoids levels[38-44].

PROGRAMMING OF INSULIN RESISTANCE, OBESITY, AND TYPE II DIABETES

Insulin resistance may come from fetal adaptation to an adverse intrauterine environment during a critical period, thus leading to programming of fetal gene expression[45,46]. Insulin plays a central role in fetal growth. During the first two years of life SGA newborns are usually able to catch-up growth by increasing their growth velocity and recovering the weight of AGA counterparts[47]. The dynamic changes that occur during this period suggest a critical role of adipose tissue in the development of metabolic complications. Ibáñez et al[48] stated that this early growth, in SGA newborns, was associated with development of central adiposity and insulin resistance between 2 to 4 years of age. The same correlation was found in early adulthood by Leunissen et al[49]. Following these epidemiological data, MetS was renamed as “the small baby syndrome”[50]. This fitted well with Hertfordshire’s findings according to which the highest risk of cardio-metabolic diseases was in men and women who had evidence of early-life deprivation (considering weight at birth or in early childhood) and who had become overweight as adults (“small becoming big”)[51]. However, we currently known that not only those subjects born with low birth weight, but also poor maternal nutrition increase maternal weight gain[52,53] and that large-for-gestational age newborns have increased metabolic risks[54].

Not only are diabetic mothers hyperglycaemic but they also have elevated circulating lipids and amino-acid. The fetal pancreas and liver are stimulated to secrete increased insulin and insulin-like growth factors that are growth-promoting hormones in the fetus. This results in the well-described diabetic mother’s macrosomic infant. Low-grade inflammation has been reported to be a link between insulin resistance, obesity, and type 2 diabetes[55]. Adipokines and cytokines affect insulin sensitivity through their ability to interfere with insulin signaling[56]; these molecules also modulate inflammation[57]. Adiponectin, which is produced by the enhanced adipose tissue, acts as insulin-sensitizing, antiatherogenic, and anti-inflammatory hormone[58]. Some scholar have shown that women with gestational diabetes mellitus (GDM) express a decreased concentration of adiponectin and an increased level of TNF-α and IL-6[57,59]. Lihn et al[60] suggest that this happens due to TNF-α and IL-6 downregulation of adiponectin expression. Leptin, which is a hormone produced by placenta and by adipocytes principally[61], is involved in weight gain regulation by interacting with neuropeptide-Y in the hypothalamus[62]. Beyond its properties as appetite-suppressant agent, Leptin is also capable of regulating lipid metabolism. Atègbo et al[57] have shown high leptin level in mothers with GDM and, in contrast, a reduced level of leptin in their macrosomic children. Leptin, as pro-inflammatory factor, may contribute to the inflammatory state during gestational diabetes. Conversely, low leptin level in macrosomic babies may contribute to weight gain since leptin-deficient rodents[62] and human[63] have been shown to develop obesity. According to the hypothesis of “Metabolic Memory”, these alterations may permanently increase the risk of trend in high food taking, overweight, obesity, and diabetogenic status in offspring during adult life[19]. An example of metabolic memory is revealed by Franke et al[64] who have shown that diabetic pregnancy in rats alters the differentiation of the newborns’ hypothalamic neurons. The impairment of these neurons may be avoided by normalizing glycemia among diabetic pregnant rats[64]. This metabolic imprinting could generate an inter-generational effect in which children risk becoming overweight or obese post-natally. Furthermore, if the child is female, she risks becoming diabetic during pregnancy, thus exposing the fetus to another route of later metabolic risk[19].

PROGRAMMING OF HYPERTENSION AND CARDIO VASCULAR DISEASE

Experimental models of fetal programming induced by gestational protein restriction[65,66], maternal stress[67], hypoxia[68] or placental insufficiency[69] demonstrate that vascular dysfunction and hypertension are related to a marked increase in glucocorticoid (GC) expression and/or marked decrease in the expression of 11β-HSD2. In these studies, the exposure to exogenous GCs generates a reduction in nephron number[70], vascular dysfunction[71], alterations in the renin-angiotensin system (RAS)[72], disruption in hypothalamic-pituitary-adrenal (HPA) axis[73-76], and hypertension[77,78] in the litter. Reduction in nephron number may affect the renal excretory function, thus contributing to the fetal programming of hypertension. However, some models demonstrate that a decrease in nephron number is sensitive to the timing of the insult[77,79] and the early-mid nephrogenesis phase is the most critical window to promote the modification in fetal kidney[80]. This change in phenotype may alter the mechanisms of adaptation to renal damage in adult life[81,82]. Otherwise other systems, which are critical to the long-term control of blood pressure, may contribute to program hypertension. As is clearly known, vascular dysfunction is implicated in the pathophysiology of hypertension[83] and plays a critical role in the development of cardio-vascular (CV) disease[84]. Many clinical studies have observed an impaired vascular function in healthy children with low birth weight[85,86], thus suggesting that vascular consequences of fetal programming may precede the development of adult CV disease. Vascular endothelial cell play a pivotal role in CV system by producing a collection of vasoactive agents whose functions include vasodilatation, vasoconstriction, and vascular growth[86]. This axiom is confirmed by animal models in which fetal insult, which is induced by nutritional restriction, placental insufficiency or hypoxia, leads to vascular dysfunction due to the impairment of endothelium-dependent nitric oxide (NO) availability[87-89]. During hypoxia, an imbalance in potent vasoactive factors is generated and an increase in total peripheral resistance is programmed, thus contributing to the development of hypertension. The RAS is another system strongly involved in blood pressure regulation and CV disease programming[90]. In the rat, RAS blockage during the nephrogenic period leads to a marked reduction in nephron number[91,92]. Although suppression of the RAS is observed at birth, hypertension is established by inappropriate activation of the RAS later in life[93-95]. According to the thrifty phenotype hypothesis, blood flow redistribution to critical organs such as the brain and heart occurs at the expense of other organs such as the liver, kidney, muscles and skin, thus resulting in exposure to hypoxia, with modifications in the hypoxia inducible factor (HIF) pathway[21]. HIF regulates several pathways, including the sympathetic nervous system, via stimulation of tyrosine hydroxylase[96]. Numerous models of fetal programming confirmed an increased amount of circulating catecholamines during placental insufficiency and gestational protein restriction[97-99]. The data are supported by the evidence that renal denervation delays the development of hypertension in prepubertal offspring[100] and abolishes hypertension in adult male IUGR offspring[101]. All these alterations in phenotype appear to contribute to hypertension in response to certain fetal insults, thus highlighting the complexity of the pathways involved in the fetal programming of hypertension and CV disease.

OS FETAL PROGRAMMING HYPOTHESIS

OS occurs when the production of free radicals (FRs) exceeds the capacity of antioxidant defenses[102]. It represents an imbalance between the production of reactive species and the capacity of biological system to readily detoxify the reactive intermediates or repair the resulting damage.

FRs can be produced through many processes. FR are generated primarily within the mitochondrial respiratory chain, which is fundamental for ATP production in mammalian cells. During the respiratory process, oxygen (O2) is utilized as an electron acceptor and completely reduced to water through the acquisition of four electrons. Once this process is completed through subsequent steps, radical formation becomes possible. NO can be also a FR source because it contains an unpaired electron in the outer orbital.

Nitric oxide synthase (NOS) catalyzes the formation of NO. It reacts relatively slowly with O2 thus producing the orange-brown gas nitrogen dioxide (.NO2), a highly reactive FR[103]. Hypoxia-ischemia sets in motion several pathways involving intracellular calcium release and activation of nitric oxide synthetase leading to increased FR generation[104].

Other potential endogenous sources of FRs include inflammatory cell activation (through Nicotinamide Adenine Dinucleotide Phosphate Reduced oxidase of phagocytes and some endothelial cells), monoxygenase system, nitric oxide synthase, and several other enzymes involved in the inflammatory process[105]. The burden of FR can be further amplified by the presence of “free” metals such as iron, copper, and manganese that are released from metalloprotein complexes[106]. Iron, can damage tissues by catalyzing the conversion of superoxide and hydrogen peroxide to FR species through the Haber-Weiss and Fenton reactions when it is unbound to plasma proteins[107].

Additional endogenous sources of cellular FR are activated neutrophils, eosinophils, and macrophages[108]. Notwithstanding the source of FRs, they are really dangerous because of their toxic effects that are able to damage all cell components, including proteins, lipids and DNA. OS may operate directly through the modulation of gene expression or indirectly through the adverse effects of oxidized molecules at critical developmental windows.

Therefore, OS causes a very wide spectrum of genetic, metabolic, and cellular responses and many oxidative conditions are able to modulate gene expression, stimulate cell growth or cause a protective temporary growth-arrest[109]. Necrosis is the most extreme outcome and involves direct cell destruction.

Recently, Leal et al[110] have shown that there is a change in the prooxidant and antioxidant defences strictly related to pregnancy process. During pregnancy, OS plays a major role in maternal-fetal interface insofar as it is essential for embryo and tissue development. Maternal diabetes, prenatal hypoxic/ischaemic events, inflammatory/infective insults are specific triggers for an acute increase in FRs, thus generating an adverse intrauterine environment with impaired fetal development[111,112]. Pro-OS is also a common feature for adverse (poor or excessive) fetal growth, preterm birth, smoking, malnutrition, overnutrition, infection and inflammation[113-116]. Consequently, OS may be the key link underlying the programming associations between adverse fetal growth/preterm birth and elevated risks of chronic diseases.

The role of OS in the pathogenesis of insulin dependent diabetes mellitus has been implicated in several studies[117,118] and there is evidence that both free-radical production and antioxidant defences are disturbed in Diabetes[119]. Hyperglycemia leads to an increased production of FRs through different metabolic pathways. In short, hyperglycemia increases formation of advanced glycation end product (AGE) and activates the hexosamine biosynthetic pathway, thus leading to the formation of glucosamine-6-phosphate that competes with glucose-6-phosphase dehydrogenase and limits the synthesis of nicotinamide adenine dinucleotide (NAD). As is clearly known, NAD is necessary for reduced glutathione (GSH) rebuilding. Moreover, activation of the polyol and protein kinase C pathways, together with oxidases activation, may also be responsible for increased FRs production[120]. Hence, end products of abnormal glucose metabolism lead to an increased formation of FRs. When FRs production overcomes fetal and placental antioxidant capacity, transcription factors (TFs) such as nuclear factor-κB, activator protein-1, and HIF-1 are activated and lead to insulin resistance due to the phosphorylation (inactivation) of insulin receptor substrate-1 (IRS-1). Inhibition of IRS-1 leads to reduced membrane translocation of glucose transport protein as glucose transporter-4 (GLUT-4), thus generating a reduction of glucose insulin-dependent up-take. Moreover, FRs are able to down-regulates GLUT-4 transcription directly[120]. Consequently, extracellular hyperglycemia occurs. However, glucose can enter all cells virtually through insulin-independent GLUTs such as GLUT-1 and GLUT-3. This raises intracellular glucose concentration and enhances FRs generation, which, again, impairs insulin and signals the establishment of a vicious circle. TFs may also directly induce the expression of pro-inflammatory cytokines such as interleukin-6, tumor necrosis factor-α or monocyte chemoattractant protein-1 that will cause insulin resistance. Recent studies in animal models have observed that manipulating anti/pro- oxidant balance in pregnancy could alter blood pressure and vascular reactivity in rat offspring[121,122]. Such emerging evidence confirms that both the insulin functional axis and blood pressure could be sensitive targets to OS programming.

OS has been demonstrated in pregnancies with fetal growth restriction[123]. Fetal growth restriction is often complicated by intrauterine hypoxia and impaired blood flow to the fetus. Intrauterine hypoxia may induce FRs generation and fetal OS. It has been demonstrated that increased isoprostanes concentrations, which are reliable markers of lipid peroxidation in amniotic fluid, indicate fetal growth restriction and also induce damage to amniotic epithelium and chorioamniotic collagen. This aspect is clarified by recent data demonstrating that F2-isoprostanes concentrations are significantly higher in pregnancies with premature rupture of membranes than in normal ones[123]. FRs may disrupt amino acid binding in proteins and polyunsaturated fatty acids of the membrane lipid bilayers, thus causing cell dysfunction, modification of chorioamniotic biology and predisposition to premature rupture of membranes.

By favouring intracellular release of NPBI into plasma, asphyxia and acidosis supply redox-cycling iron, thus predisposing to OS[124-127]. NPBI leads to the catalysis of superoxide anion (O2-.), hydrogen peroxide (H2O2), and the generation of the damaging hydroxyl radical (.OH). In presence of free iron, huge increases in FRs generation are possible and likely to cause tissue damage. Plasma NPBI may leak into the brain through a damaged barrier and is particularly damaging insofar as it is taken up by cells directly. When NPBI gains access to the extracellular space, its uptake by cells is enhanced by intracellular calcium and paradoxically also by increased levels of intracellular iron. Differentiating oligodendrocytes are particularly vulnerable to FRs damage because they are rich in iron, which is required for differentiation[128].

A recent in vivo and ex vivo rat model of IUGR underlines that delays in oligodendrocyte differentiation and myelination are probably due to bone morphogenetic protein 4 (BMP4) up-regulation induced by OS. When BMP4 expression in oligodendrocyte increases, impaired differentiation occurs. A normal myelination has been observed abrogating BMP signaling[129].

Down syndrome comes from an exceeding chromosome 21 in cellular karyotype. Superoxide dismutase (SOD) gene is localized on chromosome 21. This enzyme has the capacity to detoxify cells from superoxide anion in vivo with the participation of catalase and glutathione peroxidase. Consequently increased SOD production leads to high H2O2 generation, which can itself be toxic and also interfere with SOD activity[130]. An increased level of 8-iso-PGF2a isoprostane, was found in amniotic fluid of pregnancies with a Down syndrome foetus[131]. The immature oligodendroglial cells are glutathion peroxidase and catalase deficient so overexpression of SOD can be dangerous, instead of being protective. The early occurrence of OS in pregnancies with trisomy 21 and their subsequent oxidative damage as major contributing factor in brain aging and cognitive function decline are probably due to the overexpression of SOD, which comes from the supernumerary chromosome. SOD is also overexpressed in the immature brain, especially under stressful conditions (such as hypoxia)[132].

CONCLUSION

During early life, many gestational conditions may represent an important determinant of future health. Whereas the dominant focus of experimental studies to date has been on defining the phenotypic consequences of fetal-placental perturbations, the emphasis has now shifted to determining those initiating mechanisms underlying the programming process. The size and scope of this field has grown to include OS as the lowest common denominator. During normal pregnancies, oxidants have many physiological functions, which promote and control cellular fate and which play a crucial role in normal development through cellular signalling. In absence of a parallel increase in antioxidative activity, OS will result. Overproduction of reactive oxygen species can lead to massive cellular damage by acting on proteins, lipids, and DNA. This unbalance may change the course of pregnancy and generate a cascade effect that leads to the genesis of in utero programming of adult diseases. It is clear that placenta is not simply a passive participant in pregnancy supplying maternal substrates to the fetus. It adapts to the maternal environment and changes both its structure and function. Placenta thus assumes an active role in programming the fetal experience in utero that leads to disease in adult life. Since placenta serves as barrier against oxidative insult to maintain the homeostasis of fetal intrauterine environment, it is plausibly that placenta adaptation occurred in response to such altered maternal environment may be the general underlying mechanism that links altered placental function to fetal programming. It can also been hypothesized that programming process is extended in early postnatal life for premature infants. Premature neonates experience a hyperoxic challenge as they have to grow up in an oxygen-rich environment postnatally. Moreover, these biological systems are prone to oxidative insults because of their resilience and maturity stage at the time of insult. There could be a different timing of insult, plausibly prenatal and early postnatal periods are the most critical ‘‘windows’’ to OS programming insults.

The challenge for the future is to develop new effective antioxidant therapies and to demonstrate their benefits in treatments. However, whether antioxidant supplementation, or a diet rich in antioxidants, can avoid consequences of OS programming in the offspring or not is yet to be elucidated. Longitudinal studies evaluating the panel of OS biomarkers and elucidating the molecular mechanisms that engender OS in perinatal period are needed before antioxidant therapies are accepted in clinical practice.

Footnotes

P- Reviewer: Velasco I, Xiao DL S- Editor: Kong JX L- Editor: A E- Editor: Wang CH

References
1.  Kaur J. A comprehensive review on metabolic syndrome. Cardiol Res Pract. 2014;2014:943162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1063]  [Cited by in F6Publishing: 1053]  [Article Influence: 105.3]  [Reference Citation Analysis (0)]
2.  Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, Gordon DJ, Krauss RM, Savage PJ, Smith SC. Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute scientific statement. Curr Opin Cardiol. 2006;21:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 301]  [Cited by in F6Publishing: 306]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
3.  Lemieux I, Pascot A, Couillard C, Lamarche B, Tchernof A, Alméras N, Bergeron J, Gaudet D, Tremblay G, Prud’homme D. Hypertriglyceridemic waist: A marker of the atherogenic metabolic triad (hyperinsulinemia; hyperapolipoprotein B; small, dense LDL) in men? Circulation. 2000;102:179-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 647]  [Cited by in F6Publishing: 625]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
4.  Park YW, Zhu S, Palaniappan L, Heshka S, Carnethon MR, Heymsfield SB. The metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988-1994. Arch Intern Med. 2003;163:427-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1523]  [Cited by in F6Publishing: 1449]  [Article Influence: 69.0]  [Reference Citation Analysis (0)]
5.  Carr DB, Utzschneider KM, Hull RL, Kodama K, Retzlaff BM, Brunzell JD, Shofer JB, Fish BE, Knopp RH, Kahn SE. Intra-abdominal fat is a major determinant of the National Cholesterol Education Program Adult Treatment Panel III criteria for the metabolic syndrome. Diabetes. 2004;53:2087-2094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 638]  [Cited by in F6Publishing: 674]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
6.  Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes. 1988;37:1595-1607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5710]  [Cited by in F6Publishing: 5747]  [Article Influence: 159.6]  [Reference Citation Analysis (0)]
7.  Ferrannini E, Haffner SM, Mitchell BD, Stern MP. Hyperinsulinaemia: the key feature of a cardiovascular and metabolic syndrome. Diabetologia. 1991;34:416-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 613]  [Cited by in F6Publishing: 645]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
8.  Gustat J, Srinivasan SR, Elkasabany A, Berenson GS. Relation of self-rated measures of physical activity to multiple risk factors of insulin resistance syndrome in young adults: the Bogalusa Heart Study. J Clin Epidemiol. 2002;55:997-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 82]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
9.  Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA. 2002;287:356-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4680]  [Cited by in F6Publishing: 4417]  [Article Influence: 200.8]  [Reference Citation Analysis (0)]
10.  Apridonidze T, Essah PA, Iuorno MJ, Nestler JE. Prevalence and characteristics of the metabolic syndrome in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2005;90:1929-1935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 616]  [Cited by in F6Publishing: 544]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
11.  Elford J, Whincup P, Shaper AG. Early life experience and adult cardiovascular disease: longitudinal and case-control studies. Int J Epidemiol. 1991;20:833-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 67]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
12.  Hemachandra AH, Howards PP, Furth SL, Klebanoff MA. Birth weight, postnatal growth, and risk for high blood pressure at 7 years of age: results from the Collaborative Perinatal Project. Pediatrics. 2007;119:e1264-e1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 97]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
13.  Taylor SJ, Whincup PH, Cook DG, Papacosta O, Walker M. Size at birth and blood pressure: cross sectional study in 8-11 year old children. BMJ. 1997;314:475-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 95]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
14.  Gamborg M, Byberg L, Rasmussen F, Andersen PK, Baker JL, Bengtsson C, Canoy D, Drøyvold W, Eriksson JG, Forsén T. Birth weight and systolic blood pressure in adolescence and adulthood: meta-regression analysis of sex- and age-specific results from 20 Nordic studies. Am J Epidemiol. 2007;166:634-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 140]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
15.  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;360:659-665.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Barker DJP Mothers, babies and health in later life. London: Churchill Living-stone 1998; .  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Hales CN, Barker DJ. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. 1992. Int J Epidemiol. 2013;42:1215-1222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 162]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
18.  Godfrey KM. Maternal regulation of fetal development and health in adult life. Eur J Obstet Gynecol Reprod Biol. 1998;78:141-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 109]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
19.  Yessoufou A, Moutairou K. Maternal diabetes in pregnancy: early and long-term outcomes on the offspring and the concept of “metabolic memory”. Exp Diabetes Res. 2011;2011:218598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 165]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
20.  Bruce KD, Hanson MA. The developmental origins, mechanisms, and implications of metabolic syndrome. J Nutr. 2010;140:648-652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 181]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
21.  McMillen IC, Robinson JS. Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev. 2005;85:571-633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1352]  [Cited by in F6Publishing: 1280]  [Article Influence: 67.4]  [Reference Citation Analysis (0)]
22.  Vonnahme KA, Ford SP. Placental vascular endothelial growth factor receptor system mRNA expression in pigs selected for placental efficiency. J Physiol. 2004;554:194-201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
23.  Wallace JM, Aitken RP, Milne JS, Hay WW. Nutritionally mediated placental growth restriction in the growing adolescent: consequences for the fetus. Biol Reprod. 2004;71:1055-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 87]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
24.  Myatt L. Placental adaptive responses and fetal programming. J Physiol. 2006;572:25-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 347]  [Cited by in F6Publishing: 351]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
25.  Krebs C, Macara LM, Leiser R, Bowman AW, Greer IA, Kingdom JC. Intrauterine growth restriction with absent end-diastolic flow velocity in the umbilical artery is associated with maldevelopment of the placental terminal villous tree. Am J Obstet Gynecol. 1996;175:1534-1542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 317]  [Cited by in F6Publishing: 254]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
26.  Jansson T, Ekstrand Y, Björn C, Wennergren M, Powell TL. Alterations in the activity of placental amino acid transporters in pregnancies complicated by diabetes. Diabetes. 2002;51:2214-2219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 159]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
27.  McMullen S, Osgerby JC, Thurston LM, Gadd TS, Wood PJ, Wathes DC, Michael AE. Alterations in placental 11 beta-hydroxysteroid dehydrogenase (11 betaHSD) activities and fetal cortisol: cortisone ratios induced by nutritional restriction prior to conception and at defined stages of gestation in ewes. Reproduction. 2004;127:717-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
28.  Jansson N, Greenwood SL, Johansson BR, Powell TL, Jansson T. Leptin stimulates the activity of the system A amino acid transporter in human placental villous fragments. J Clin Endocrinol Metab. 2003;88:1205-1211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 166]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
29.  Mahendran D, Donnai P, Glazier JD, D’Souza SW, Boyd RD, Sibley CP. Amino acid (system A) transporter activity in microvillous membrane vesicles from the placentas of appropriate and small for gestational age babies. Pediatr Res. 1993;34:661-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 166]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
30.  Ayuk PT, Theophanous D, D’Souza SW, Sibley CP, Glazier JD. L-arginine transport by the microvillous plasma membrane of the syncytiotrophoblast from human placenta in relation to nitric oxide production: effects of gestation, preeclampsia, and intrauterine growth restriction. J Clin Endocrinol Metab. 2002;87:747-751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
31.  Cramer S, Beveridge M, Kilberg M, Novak D. Physiological importance of system A-mediated amino acid transport to rat fetal development. Am J Physiol Cell Physiol. 2002;282:C153-C160.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Lindsay RS, Lindsay RM, Waddell BJ, Seckl JR. Prenatal glucocorticoid exposure leads to offspring hyperglycaemia in the rat: studies with the 11 beta-hydroxysteroid dehydrogenase inhibitor carbenoxolone. Diabetologia. 1996;39:1299-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 224]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
33.  Welberg LA, Seckl JR, Holmes MC. Inhibition of 11beta-hydroxysteroid dehydrogenase, the foeto-placental barrier to maternal glucocorticoids, permanently programs amygdala GR mRNA expression and anxiety-like behaviour in the offspring. Eur J Neurosci. 2000;12:1047-1054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 247]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
34.  Krozowski Z, MaGuire JA, Stein-Oakley AN, Dowling J, Smith RE, Andrews RK. Immunohistochemical localization of the 11 beta-hydroxysteroid dehydrogenase type II enzyme in human kidney and placenta. J Clin Endocrinol Metab. 1995;80:2203-2209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
35.  Seckl JR, Cleasby M, Nyirenda MJ. Glucocorticoids, 11beta-hydroxysteroid dehydrogenase, and fetal programming. Kidney Int. 2000;57:1412-1417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 188]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
36.  Jauniaux E, Watson AL, Hempstock J, Bao YP, Skepper JN, Burton GJ. Onset of maternal arterial blood flow and placental oxidative stress. A possible factor in human early pregnancy failure. Am J Pathol. 2000;157:2111-2122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 776]  [Cited by in F6Publishing: 698]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
37.  Wang Y, Walsh SW, Kay HH. Placental lipid peroxides and thromboxane are increased and prostacyclin is decreased in women with preeclampsia. Am J Obstet Gynecol. 1992;167:946-949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 157]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
38.  Alexander BT. Placental insufficiency leads to development of hypertension in growth-restricted offspring. Hypertension. 2003;41:457-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 235]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
39.  Edwards LJ, Coulter CL, Symonds ME, McMillen IC. Prenatal undernutrition, glucocorticoids and the programming of adult hypertension. Clin Exp Pharmacol Physiol. 2001;28:938-941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 65]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
40.  Langley-Evans SC. Intrauterine programming of hypertension by glucocorticoids. Life Sci. 1997;60:1213-1221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 73]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
41.  Nathanielsz PW. Animal models that elucidate basic principles of the developmental origins of adult diseases. ILAR J. 2006;47:73-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 136]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
42.  Ross MG, Desai M, Guerra C, Wang S. Programmed syndrome of hypernatremic hypertension in ovine twin lambs. Am J Obstet Gynecol. 2005;192:1196-1204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
43.  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;49:460-467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 458]  [Cited by in F6Publishing: 402]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
44.  Vehaskari VM, Woods LL. Prenatal programming of hypertension: lessons from experimental models. J Am Soc Nephrol. 2005;16:2545-2556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
45.  Mericq V, Ong KK, Bazaes R, Peña V, Avila A, Salazar T, Soto N, Iñiguez G, Dunger DB. Longitudinal changes in insulin sensitivity and secretion from birth to age three years in small- and appropriate-for-gestational-age children. Diabetologia. 2005;48:2609-2614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 214]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
46.  Ford ES, Li C. Defining the metabolic syndrome in children and adolescents: will the real definition please stand up? J Pediatr. 2008;152:160-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 218]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
47.  Boersma B, Wit JM. Catch-up growth. Endocr Rev. 1997;18:646-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 140]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
48.  Ibáñez L, Ong K, Dunger DB, de Zegher F. Early development of adiposity and insulin resistance after catch-up weight gain in small-for-gestational-age children. J Clin Endocrinol Metab. 2006;91:2153-2158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 384]  [Cited by in F6Publishing: 367]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
49.  Leunissen RW, Kerkhof GF, Stijnen T, Hokken-Koelega A. Timing and tempo of first-year rapid growth in relation to cardiovascular and metabolic risk profile in early adulthood. JAMA. 2009;301:2234-2242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 361]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
50.  Barker DJ, Hales CN, Fall CH, Osmond C, Phipps K, Clark PM. Type 2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): relation to reduced fetal growth. Diabetologia. 1993;36:62-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1710]  [Cited by in F6Publishing: 1574]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
51.  Fall CH. Fetal programming and the risk of noncommunicable disease. Indian J Pediatr. 2013;80 Suppl 1:S13-S20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
52.  Fraser A, Tilling K, Macdonald-Wallis C, Sattar N, Brion MJ, Benfield L, Ness A, Deanfield J, Hingorani A, Nelson SM. Association of maternal weight gain in pregnancy with offspring obesity and metabolic and vascular traits in childhood. Circulation. 2010;121:2557-2564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 383]  [Cited by in F6Publishing: 343]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
53.  Reynolds RM, Osmond C, Phillips DI, Godfrey KM. Maternal BMI, parity, and pregnancy weight gain: influences on offspring adiposity in young adulthood. J Clin Endocrinol Metab. 2010;95:5365-5369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 176]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
54.  Dyer JS, Rosenfeld CR, Rice J, Rice M, Hardin DS. Insulin resistance in Hispanic large-for-gestational-age neonates at birth. J Clin Endocrinol Metab. 2007;92:3836-3843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
55.  Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol. 2004;25:4-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1432]  [Cited by in F6Publishing: 1459]  [Article Influence: 73.0]  [Reference Citation Analysis (0)]
56.  Greenberg AS, McDaniel ML. Identifying the links between obesity, insulin resistance and beta-cell function: potential role of adipocyte-derived cytokines in the pathogenesis of type 2 diabetes. Eur J Clin Invest. 2002;32 Suppl 3:24-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 204]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
57.  Atègbo JM, Grissa O, Yessoufou A, Hichami A, Dramane KL, Moutairou K, Miled A, Grissa A, Jerbi M, Tabka Z. Modulation of adipokines and cytokines in gestational diabetes and macrosomia. J Clin Endocrinol Metab. 2006;91:4137-4143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 265]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
58.  Díez JJ, Iglesias P. The role of the novel adipocyte-derived hormone adiponectin in human disease. Eur J Endocrinol. 2003;148:293-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 759]  [Cited by in F6Publishing: 742]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
59.  Meller M, Qiu C, Vadachkoria S, Abetew DF, Luthy DA, Williams MA. Changes in placental adipocytokine gene expression associated with gestational diabetes mellitus. Physiol Res. 2006;55:501-512.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Lihn AS, Richelsen B, Pedersen SB, Haugaard SB, Rathje GS, Madsbad S, Andersen O. Increased expression of TNF-alpha, IL-6, and IL-8 in HALS: implications for reduced adiponectin expression and plasma levels. Am J Physiol Endocrinol Metab. 2003;285:E1072-E1080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 135]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
61.  Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425-432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9119]  [Cited by in F6Publishing: 8625]  [Article Influence: 287.5]  [Reference Citation Analysis (0)]
62.  Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, Lallone RL, Burley SK, Friedman JM. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995;269:543-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3184]  [Cited by in F6Publishing: 3014]  [Article Influence: 103.9]  [Reference Citation Analysis (0)]
63.  Montague CT, Prins JB, Sanders L, Zhang J, Sewter CP, Digby J, Byrne CD, O’Rahilly S. Depot-related gene expression in human subcutaneous and omental adipocytes. Diabetes. 1998;47:1384-1391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 249]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
64.  Franke K, Harder T, Aerts L, Melchior K, Fahrenkrog S, Rodekamp E, Ziska T, Van Assche FA, Dudenhausen JW, Plagemann A. ‘Programming’ of orexigenic and anorexigenic hypothalamic neurons in offspring of treated and untreated diabetic mother rats. Brain Res. 2005;1031:276-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 117]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
65.  Langley-Evans SC, Phillips GJ, Benediktsson R, Gardner DS, Edwards CR, Jackson AA, Seckl JR. Protein intake in pregnancy, placental glucocorticoid metabolism and the programming of hypertension in the rat. Placenta. 1996;17:169-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 302]  [Cited by in F6Publishing: 314]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
66.  Bertram C, Trowern AR, Copin N, Jackson AA, Whorwood CB. The maternal diet during pregnancy programs altered expression of the glucocorticoid receptor and type 2 11beta-hydroxysteroid dehydrogenase: potential molecular mechanisms underlying the programming of hypertension in utero. Endocrinology. 2001;142:2841-2853.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
67.  Takahashi LK, Turner JG, Kalin NH. Prolonged stress-induced elevation in plasma corticosterone during pregnancy in the rat: implications for prenatal stress studies. Psychoneuroendocrinology. 1998;23:571-581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 136]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
68.  Hardy DB, Yang K. The expression of 11 beta-hydroxysteroid dehydrogenase type 2 is induced during trophoblast differentiation: effects of hypoxia. J Clin Endocrinol Metab. 2002;87:3696-3701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
69.  Baserga M, Hale MA, Wang ZM, Yu X, Callaway CW, McKnight RA, Lane RH. Uteroplacental insufficiency alters nephrogenesis and downregulates cyclooxygenase-2 expression in a model of IUGR with adult-onset hypertension. Am J Physiol Regul Integr Comp Physiol. 2007;292:R1943-R1955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
70.  Wintour EM, Moritz KM, Johnson K, Ricardo S, Samuel CS, Dodic M. Reduced nephron number in adult sheep, hypertensive as a result of prenatal glucocorticoid treatment. J Physiol. 2003;549:929-935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 198]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
71.  Hadoke PW, Lindsay RS, Seckl JR, Walker BR, Kenyon CJ. Altered vascular contractility in adult female rats with hypertension programmed by prenatal glucocorticoid exposure. J Endocrinol. 2006;188:435-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
72.  Moritz KM, Johnson K, Douglas-Denton R, Wintour EM, Dodic M. Maternal glucocorticoid treatment programs alterations in the renin-angiotensin system of the ovine fetal kidney. Endocrinology. 2002;143:4455-4463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 100]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
73.  Ortiz LA, Quan A, Zarzar F, Weinberg A, Baum M. Prenatal dexamethasone programs hypertension and renal injury in the rat. Hypertension. 2003;41:328-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 200]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
74.  Kapoor A, Petropoulos S, Matthews SG. Fetal programming of hypothalamic-pituitary-adrenal (HPA) axis function and behavior by synthetic glucocorticoids. Brain Res Rev. 2008;57:586-595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 188]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
75.  O‘Regan D, Welberg LL, Holmes MC, Seckl JR. Glucocorticoid programming of pituitary-adrenal function: mechanisms and physiological consequences. Semin Neonatol. 2001;6:319-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
76.  Benediktsson R, Lindsay RS, Noble J, Seckl JR, Edwards CR. Glucocorticoid exposure in utero: new model for adult hypertension. Lancet. 1993;341:339-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 607]  [Cited by in F6Publishing: 565]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
77.  Dodic M, Abouantoun T, O’Connor A, Wintour EM, Moritz KM. Programming effects of short prenatal exposure to dexamethasone in sheep. Hypertension. 2002;40:729-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 153]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
78.  Roghair RD, Lamb FS, Miller FJ, Scholz TD, Segar JL. Early gestation dexamethasone programs enhanced postnatal ovine coronary artery vascular reactivity. Am J Physiol Regul Integr Comp Physiol. 2005;288:R46-R53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
79.  Woods LL, Weeks DA, Rasch R. Programming of adult blood pressure by maternal protein restriction: role of nephrogenesis. Kidney Int. 2004;65:1339-1348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 266]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
80.  Guron G, Friberg P. An intact renin-angiotensin system is a prerequisite for normal renal development. J Hypertens. 2000;18:123-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 172]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
81.  Bursztyn M, Gross ML, Goltser-Dubner T, Koleganova N, Birman T, Smith Y, Ariel I. Adult hypertension in intrauterine growth-restricted offspring of hyperinsulinemic rats: evidence of subtle renal damage. Hypertension. 2006;48:717-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
82.  Nwagwu MO, Cook A, Langley-Evans SC. Evidence of progressive deterioration of renal function in rats exposed to a maternal low-protein diet in utero. Br J Nutr. 2000;83:79-85.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Christensen KL, Mulvany MJ. Location of resistance arteries. J Vasc Res. 2001;38:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 163]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
84.  Panza JA, Quyyumi AA, Brush JE, Epstein SE. Abnormal endothelium-dependent vascular relaxation in patients with essential hypertension. N Engl J Med. 1990;323:22-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1763]  [Cited by in F6Publishing: 1812]  [Article Influence: 53.3]  [Reference Citation Analysis (0)]
85.  Martin H, Hu J, Gennser G, Norman M. Impaired endothelial function and increased carotid stiffness in 9-year-old children with low birthweight. Circulation. 2000;102:2739-2744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 213]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
86.  Beevers G, Lip GY, O’Brien E. ABC of hypertension: The pathophysiology of hypertension. BMJ. 2001;322:912-916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 217]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
87.  Payne JA, Alexander BT, Khalil RA. Reduced endothelial vascular relaxation in growth-restricted offspring of pregnant rats with reduced uterine perfusion. Hypertension. 2003;42:768-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
88.  Williams SJ, Hemmings DG, Mitchell JM, McMillen IC, Davidge ST. Effects of maternal hypoxia or nutrient restriction during pregnancy on endothelial function in adult male rat offspring. J Physiol. 2005;565:125-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 99]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
89.  Moritz KM, Dodic M, Wintour EM. Kidney development and the fetal programming of adult disease. Bioessays. 2003;25:212-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 103]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
90.  Hall JE, Brands MW, Henegar JR. Angiotensin II and long-term arterial pressure regulation: the overriding dominance of the kidney. J Am Soc Nephrol. 1999;10 Suppl 12:S258-S265.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Rasch R, Skriver E, Woods LL. The role of the RAS in programming of adult hypertension. Acta Physiol Scand. 2004;181:537-542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
92.  Saez F, Castells MT, Zuasti A, Salazar F, Reverte V, Loria A, Salazar FJ. Sex differences in the renal changes elicited by angiotensin II blockade during the nephrogenic period. Hypertension. 2007;49:1429-1435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
93.  Grigore D, Ojeda NB, Robertson EB, Dawson AS, Huffman CA, Bourassa EA, Speth RC, Brosnihan KB, Alexander BT. Placental insufficiency results in temporal alterations in the renin angiotensin system in male hypertensive growth restricted offspring. Am J Physiol Regul Integr Comp Physiol. 2007;293:R804-R811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 91]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
94.  Manning J, Vehaskari VM. Low birth weight-associated adult hypertension in the rat. Pediatr Nephrol. 2001;16:417-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 121]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
95.  Sahajpal V, Ashton N. Renal function and angiotensin AT1 receptor expression in young rats following intrauterine exposure to a maternal low-protein diet. Clin Sci (Lond). 2003;104:607-614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 116]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
96.  Leclere N, Andreeva N, Fuchs J, Kietzmann T, Gross J. Hypoxia-induced long-term increase of dopamine and tyrosine hydroxylase mRNA levels. Prague Med Rep. 2004;105:291-300.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Hiraoka T, Kudo T, Kishimoto Y. Catecholamines in experimentally growth-retarded rat fetus. Asia Oceania J Obstet Gynaecol. 1991;17:341-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
98.  Jones CT, Robinson JS. Studies on experimental growth retardation in sheep. Plasma catecholamines in fetuses with small placenta. J Dev Physiol. 1983;5:77-87.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Petry CJ, Dorling MW, Wang CL, Pawlak DB, Ozanne SE. Catecholamine levels and receptor expression in low protein rat offspring. Diabet Med. 2000;17:848-853.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 55]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
100.  Ojeda NB, Johnson WR, Dwyer TM, Alexander BT. Early renal denervation prevents development of hypertension in growth-restricted offspring. Clin Exp Pharmacol Physiol. 2007;34:1212-1216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
101.  Alexander BT, Hendon AE, Ferril G, Dwyer TM. Renal denervation abolishes hypertension in low-birth-weight offspring from pregnant rats with reduced uterine perfusion. Hypertension. 2005;45:754-758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 125]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
102.  Buonocore G, Groenendaal F. Anti-oxidant strategies. Semin Fetal Neonatal Med. 2007;12:287-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 91]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
103.  Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J. 2001;357:593-615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1895]  [Cited by in F6Publishing: 1814]  [Article Influence: 78.9]  [Reference Citation Analysis (0)]
104.  Buonocore G, Perrone S, Bracci R. Free radicals and brain damage in the newborn. Biol Neonate. 2001;79:180-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 145]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
105.  Auten RL, Davis JM. Oxygen toxicity and reactive oxygen species: the devil is in the details. Pediatr Res. 2009;66:121-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 395]  [Cited by in F6Publishing: 409]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
106.  McCarthy SM, Bove PF, Matthews DE, Akaike T, van der Vliet A. Nitric oxide regulation of MMP-9 activation and its relationship to modifications of the cysteine switch. Biochemistry. 2008;47:5832-5840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 48]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
107.  Gutteridge JM. Fate of oxygen free radicals in extracellular fluids. Biochem Soc Trans. 1982;10:72-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
108.  Conner EM, Brand SJ, Davis JM, Kang DY, Grisham MB. Role of reactive metabolites of oxygen and nitrogen in inflammatory bowel disease: toxins, mediators, and modulators of gene expression. Inflamm Bowel Dis. 1996;2:133-147.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Davies KJ. An overview of oxidative stress. IUBMB Life. 2000;50:241-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 18]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
110.  Leal CA, Schetinger MR, Leal DB, Morsch VM, da Silva AS, Rezer JF, de Bairros AV, Jaques JA. Oxidative stress and antioxidant defenses in pregnant women. Redox Rep. 2011;16:230-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
111.  Buonocore G, Perrone S, Longini M, Terzuoli L, Bracci R. Total hydroperoxide and advanced oxidation protein products in preterm hypoxic babies. Pediatr Res. 2000;47:221-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 121]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
112.  Buonocore G, Perrone S, Longini M, Vezzosi P, Marzocchi B, Paffetti P, Bracci R. Oxidative stress in preterm neonates at birth and on the seventh day of life. Pediatr Res. 2002;52:46-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 143]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
113.  Phelps DL. Retinopathy of prematurity: an estimate of vision loss in the United States--1979. Pediatrics. 1981;67:924-925.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Cooke RW. Factors affecting survival and outcome at 3 years in extremely preterm infants. Arch Dis Child Fetal Neonatal Ed. 1994;71:F28-F31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 38]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
115.  Fanaroff AA, Wright LL, Stevenson DK, Shankaran S, Donovan EF, Ehrenkranz RA, Younes N, Korones SB, Stoll BJ, Tyson JE. Very-low-birth-weight outcomes of the National Institute of Child Health and Human Development Neonatal Research Network, May 1991 through December 1992. Am J Obstet Gynecol. 1995;173:1423-1431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 223]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
116.  Stevenson DK, Wright LL, Lemons JA, Oh W, Korones SB, Papile LA, Bauer CR, Stoll BJ, Tyson JE, Shankaran S. Very low birth weight outcomes of the National Institute of Child Health and Human Development Neonatal Research Network, January 1993 through December 1994. Am J Obstet Gynecol. 1998;179:1632-1639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 268]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
117.  Jain SK. Hyperglycemia can cause membrane lipid peroxidation and osmotic fragility in human red blood cells. J Biol Chem. 1989;264:21340-21345.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Sato Y, Hotta N, Sakamoto N, Matsuoka S, Ohishi N, Yagi K. Lipid peroxide level in plasma of diabetic patients. Biochem Med. 1979;21:104-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 308]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
119.  Lyons TJ. Oxidized low density lipoproteins: a role in the pathogenesis of atherosclerosis in diabetes? Diabet Med. 1991;8:411-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 139]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
120.  Lappas M, Andrikopoulos S, Permezel M. Hypoxanthine-xanthine oxidase down-regulates GLUT1 transcription via SIRT1 resulting in decreased glucose uptake in human placenta. J Endocrinol. 2012;213:49-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
121.  Franco Mdo C, Dantas AP, Akamine EH, Kawamoto EM, Fortes ZB, Scavone C, Tostes RC, Carvalho MH, Nigro D. Enhanced oxidative stress as a potential mechanism underlying the programming of hypertension in utero. J Cardiovasc Pharmacol. 2002;40:501-509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 107]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
122.  Racasan S, Braam B, van der Giezen DM, Goldschmeding R, Boer P, Koomans HA, Joles JA. Perinatal L-arginine and antioxidant supplements reduce adult blood pressure in spontaneously hypertensive rats. Hypertension. 2004;44:83-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 91]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
123.  Longini M, Perrone S, Kenanidis A, Vezzosi P, Marzocchi B, Petraglia F, Centini G, Buonocore G. Isoprostanes in amniotic fluid: a predictive marker for fetal growth restriction in pregnancy. Free Radic Biol Med. 2005;38:1537-1541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 61]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
124.  Buonocore G, Zani S, Perrone S, Caciotti B, Bracci R. Intraerythrocyte nonprotein-bound iron and plasma malondialdehyde in the hypoxic newborn. Free Radic Biol Med. 1998;25:766-770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
125.  Buonocore G, Perrone S. Biomarkers of hypoxic brain injury in the neonate. Clin Perinatol. 2004;31:107-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
126.  Ciccoli L, Rossi V, Leoncini S, Signorini C, Paffetti P, Bracci R, Buonocore G, Comporti M. Iron release in erythrocytes and plasma non protein-bound iron in hypoxic and non hypoxic newborns. Free Radic Res. 2003;37:51-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
127.  Comporti M, Signorini C, Buonocore G, Ciccoli L. Iron release, oxidative stress and erythrocyte ageing. Free Radic Biol Med. 2002;32:568-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 153]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
128.  Ozawa H, Nishida A, Mito T, Takashima S. Development of ferritin-containing cells in the pons and cerebellum of the human brain. Brain Dev. 1994;16:92-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
129.  Reid MV, Murray KA, Marsh ED, Golden JA, Simmons RA, Grinspan JB. Delayed myelination in an intrauterine growth retardation model is mediated by oxidative stress upregulating bone morphogenetic protein 4. J Neuropathol Exp Neurol. 2012;71:640-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
130.  Bray RC, Cockle SA, Fielden EM, Roberts PB, Rotilio G, Calabrese L. Reduction and inactivation of superoxide dismutase by hydrogen peroxide. Biochem J. 1974;139:43-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 305]  [Cited by in F6Publishing: 325]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
131.  Perrone S, Longini M, Bellieni CV, Centini G, Kenanidis A, De Marco L, Petraglia F, Buonocore G. Early oxidative stress in amniotic fluid of pregnancies with Down syndrome. Clin Biochem. 2007;40:177-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
132.  Okado-Matsumoto A, Fridovich I. Subcellular distribution of superoxide dismutases (SOD) in rat liver: Cu,Zn-SOD in mitochondria. J Biol Chem. 2001;276:38388-38393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 732]  [Cited by in F6Publishing: 712]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]