Hostname: page-component-76fb5796d-dfsvx Total loading time: 0 Render date: 2024-04-26T19:41:36.755Z Has data issue: false hasContentIssue false

Developmental programming of cardiovascular disease by prenatal hypoxia

Published online by Cambridge University Press:  18 April 2013

D. A. Giussani*
Affiliation:
Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
S. T. Davidge
Affiliation:
Departments of Obstetrics & Gynecology and Physiology, Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
*
*Address for correspondence: Prof. D. A. Giussani, Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK. (Email dag26@cam.ac.uk)
Rights & Permissions [Opens in a new window]

Abstract

It is now recognized that the quality of the fetal environment during early development is important in programming cardiovascular health and disease in later life. Fetal hypoxia is one of the most common consequences of complicated pregnancies worldwide. However, in contrast to the extensive research effort on pregnancy affected by maternal nutrition or maternal stress, the contribution of pregnancy affected by fetal chronic hypoxia to developmental programming is only recently becoming delineated and established. This review discusses the increasing body of evidence supporting the programming of cardiac susceptibility to ischaemia and reperfusion (I/R) injury, of endothelial dysfunction in peripheral resistance circulations, and of indices of the metabolic syndrome in adult offspring of hypoxic pregnancy. An additional focus of the review is the identification of plausible mechanisms and the implementation of maternal and early life interventions to protect against adverse programming.

Type
Review
Copyright
Copyright © Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2013 

Developmental programming of cardiovascular disease

Heart disease is the greatest killer in the world today, imposing a staggering burden on every nation's health and wealth.1 Worldwide, it results in one in three deaths per year. Moreover, the economic costs are tremendous for treatment, patient care and lost workforce amounting to over £30 billion per year in the United Kingdom alone2 and over $US 130 billion/year in Canada and the United States.3, Reference Heidenreich, Trogdon and Khavjou4 Therefore, there is no question that cardiovascular disease is an important as well as an expensive problem to resolve. The concept that traditional lifestyle risk factors, such as smoking and obesity, interact with our genetic makeup to determine a risk of cardiovascular disease is well accepted.Reference Agarwal, Williams and Fisher5 However, only comparatively recently, it has become appreciated that the interaction between our genes and the quality of the environment during early development may be just as, if not more, important in programming cardiovascular health and disease in later life.Reference Barker6, Reference Gluckman, Hanson, Cooper and Thornburg7 This additional concept of developmental programming of disease is supported by overwhelming evidence derived from human studies now dating back more than two decades and encompassing six continents; evidence that strongly links development under sub-optimal intrauterine conditions with fetal growth restriction, low birth weight and increased rates of coronary heart disease and the metabolic syndrome in adulthood.Reference Barker, Osmond, Winter, Margetts and Simmonds8Reference Silva, Santos and Amigo14 It is generally accepted that the more immature the individual, the greater the influence the environment will have on it. Therefore, the impact of the environment in the programming of physiology is greatest in early life and decreases from embryonic to fetal to postnatal and adult life. Similarly, the opportunity for correction of a change in this developmental programme follows a similar trajectory, drastically diminishing from early life to adulthood. Hence, the concept of programming of disease creates an exciting window of opportunity to diagnose and halt the development of heart disease at its very origin, bringing preventative medicine back into the womb, or to treat postnatally as soon as possible after diagnosis to diminish and control the progression of disease. However, the mechanisms underlying developmental origins of cardiovascular dysfunction remain elusive, precluding the identification of potential clinical therapy.

Developmental programming by different suboptimal conditions during pregnancy

Epidemiological studies relating the type of suboptimal intrauterine condition with physiological dysfunction in later life have largely focussed on human populations undergoing alterations in maternal nutrition, or on human pregnancy affected by maternal psychological stress or by exposure to stress hormones.Reference Roseboom, van der Meulen and Ravelli15Reference Dalziel, Walker and Parag17 This focus on the nutrient supply to the fetus or on materno-fetal stress in humans is supported by a large number of investigations in experimental animal models demonstrating that cardiovascular dysfunction in adulthood can be programmed in pregnancy by inappropriate nutrition or by exposure to glucocorticoid excess.Reference Gluckman, Hanson, Cooper and Thornburg7, Reference McMillen and Robinson18, Reference Seckl and Meaney19 In addition to alterations in maternal nutrition and maternal stress, fetal hypoxia is one of, if not the most common consequence of complicated pregnancy worldwide (see Koos).Reference Koos20 Further, over 140 million people live at altitudes higher than 2500 m where lowered oxygen availability has been shown to reduce fetal growth and birth weight,Reference Giussani, Phillips, Anstee and Barker21, Reference Moore, Charles and Julian22 thereby comprising the largest single human group at risk for fetal growth restriction. However, in contrast to the international research effort on pregnancy affected by nutrition or glucocorticoid exposure, the contribution of fetal chronic hypoxia to developmental programming has only recently become delineated and established.

The fetal cardiovascular defence to hypoxia

The fetal cardiovascular responses to short-term episodes of acute hypoxia include a redistribution of the cardiac output away from peripheral circulations to maintain perfusion to the brain.Reference Cohn, Sacks, Heymann and Rudolph23 This redistribution of blood flow is aided by peripheral vasoconstriction, the latter significantly also decreasing oxygen consumption in less essential vascular beds.Reference Boyle, Hirst, Zerbe, Meschia and Wilkening24 The physiology underlying this circulatory response is well established and involves the activation of a selective carotid body chemoreflex.Reference Giussani, Spencer, Moore, Bennet and Hanson25 Once initiated, the neurally triggered vascular response is maintained by the release of constrictor agents into the fetal circulation, such as catecholamines, vasopressin and neuropeptide Y.Reference Jones and Robinson26Reference Perez, Espinoza, Riquelme, Parer and Llanos28 More recently, it has become clear that these neuroendocrine constrictor influences on the fetal peripheral circulation can be further influenced by the release of local vascular agents during acute hypoxia. Studies have shown that nitric oxide (NO) bioavailability in the fetus can increase during acute hypoxia.Reference Morrison, Gardner, Fletcher, Bloomfield and Giussani29 Further, increased generation of reactive oxygen species (ROS), such as the superoxide anion (O), during acute hypoxia can interact with NO, providing an oxidant tone to the fetal vasculature.Reference Thakor, Richter and Kane30, Reference Thakor, Herrera, Serón-Ferré and Giussani31 Therefore, the fetal peripheral vasoconstrictor response to acute hypoxia is an aggregate of carotid chemoreflex activation, humoral constrictor influences and a local oxidant tone determined by the ratio of NO: O in the fetal circulation.Reference Thakor, Richter and Kane30Reference Herrera, Kane and Hansell33

There is now a considerable evidence that this homeostatic circulatory defence to hypoxia is maintained, should the duration of the hypoxic challenge persist.Reference Kamitomo, Alonso, Okai, Longo and Gilbert34Reference McMillen, Adams, Ross and Coulter37 In response to fetal chronic hypoxia, the maintained redistribution of blood flow away from peripheral circulations can become maladaptive, triggering a number of unwanted side effects in the fetus. The most described consequence is asymmetric fetal growth restriction, yielding offspring whose brain and heart growth is spared, but having bodies that are thin for their length with a low ponderal index.Reference Barker6 In addition, sustained increases in fetal peripheral vascular resistance because of sustained prenatal hypoxia will increase fetal arterial blood pressure if cardiac output is maintained. An increase in fetal cardiac afterload may trigger changes in the morphology and function of the fetal heart. In turn, remodelling of the walls of the fetal aorta may occur in response to the greater pressure generated by the fetal heart. Therefore, depending on the timing, duration and severity of the challenge, chronic fetal hypoxia is not only an immediate threat to fetal survival, but it is also an important environmental influence triggering intrauterine growth restriction (IUGR) and the developmental programming of cardiovascular disease.

Chronic hypoxia and fetal origins of cardiovascular disease

Over the years, various animal models have been created to induce adverse intrauterine conditions. Many of these techniques have involved impairing utero-placental perfusion, which reduces nutrient as well as oxygen delivery to the fetus, and these studies have been the subject of several excellent reviews.Reference McMillen and Robinson18, Reference Morrison36Reference Langley-Evans, Bellinger and McMullen41 Other studies have concentrated on determining the contribution of fetal chronic hypoxia alone in promoting fetal growth restriction and early origins of cardiac and peripheral vascular dysfunction; it is these studies that will be the focus of this review.

A cluster of research groups have employed the chick embryo model, which isolates the effects of chronic hypoxia on fetal growth and the developing cardiovascular system independent of effects on the maternal and placental physiology. It is now established that exposure of the chick embryo to chronic hypoxia promotes asymmetric fetal growth restriction, cardiac and aortic hypertrophic growth, altered cardiac function and sympathetic hyper-innervation of peripheral resistance arteries by the end of the incubation period.Reference Ruijtenbeek, le Noble and Janssen42Reference Lindgren and Altimiras53 The asymmetric growth restriction and cardiac and aortic wall remodelling that develops in sea level chick embryos incubated at high altitude no longer occurs in sea level embryos incubated at high altitude with oxygen supplementation,Reference Giussani, Salinas, Villena and Blanco49, Reference Salinas, Blanco and Villena52 underlying the direct effects of isolated chronic hypoxia on fetal growth and cardiovascular development. Asymmetric growth restriction, aortic wall thickening, cardiac and vascular dysfunction have also been reported in the chronically hypoxic fetus of mammalian species, such as in sheep, rodents and guinea pigs.Reference Jacobs, Robinson, Owens, Falconer and Webster54Reference Hemmings, Williams and Davidge73 Fetal aortic wall thickening is particularly relevant in the clinical setting, as increased large artery stiffness independently predicts cardiovascular risk in humans,Reference McEniery and Wilkinson74 being a key component in the aetiology of hypertension, atherosclerosis and coronary heart disease.Reference Arnett, Evans and Riley75 In the aorta, in particular, an increase in wall thickness has been proposed as the first physical sign in the development of atherosclerosis.Reference Crispi, Figueras and Cruz-Lemini76 Aortic pulse-wave velocity measurements, rather than systolic blood pressure measurement, better predict later cardiovascular disease, including impaired coronary artery flow and left ventricular dysfunction.Reference Cruickshank, Riste and Anderson77 A comprehensive series of studies by Gilbert and colleaguesReference Alonso, Okai, Longo and Gilbert56Reference Gilbert, Pearce and Longo64 in fetal sheep subjected to high altitude from day 30 of gestation to term (ca. 145 days) reported that in the chronically hypoxic ovine fetus, cardiac output was decreased secondary to a decrease in myocardial cell contractile function. The intracellular mechanisms responsible for these reductions included reduced myofibrillar Mg2+-activated ATPase and a decrease in β1-adrenergic receptor stimulated influence on myocardial contraction. An overproduction of cAMP by β1-adrenergic receptor stimulation, promoting over-phosphorylation of troponin I may also contribute to reduced calcium binding by troponin C. Similarly, Sharma et al.Reference Sharma, Lucitti and Nordman47 reported that chronic hypoxia in the chick embryo decreased maximum ventricular + dP/dt and peak pressure, increased ventricular end-systolic volume, and decreased ventricular ejection fraction, consistent with depressed systolic function. In the same study, it was shown that arterial after-load increased and steady-state hydraulic power decreased in response to hypoxic incubation.Reference Sharma, Lucitti and Nordman47 Four separate human clinical studiesReference Skilton, Evans, Griffiths, Harmer and Celermajer78Reference Cosmi, Visentin, Fanelli, Mautone and Zanardo81 have now reported that babies born from pregnancies complicated by placental insufficiency show aortic thickening with increased vascular stiffness and reduced distensibility. Additional reported abnormalities in cardiovascular morphology and function of the human IUGR fetus include an increase in relative heart weight and ventricular wall hypertrophy,Reference Veille, Hanson, Sivakoff, Hoen and Ben-Ami82 a decrease in ventricle and myocyte volume,Reference Mayhew, Gregson and Fagan83 and compromized biventricular ejection forceReference Rizzo, Capponi, Rinaldo, Arduini and Romanini84 and diastolic filling.Reference Miyague, Ghidini, Fromberg and Miyague85 Since fetal hypoxia alone can trigger fetal aortic wall thickening and fetal cardiac dysfunction in experimental animal models, a reasonable assumption is that fetal hypoxia alone may be responsible for triggering similar cardiovascular defects in the human fetus in pregnancies complicated by placental insufficiency.

Chronic hypoxia and programming cardiovascular disease in adulthood

With an established contribution of chronic hypoxia to a fetal origin of cardiovascular dysfunction, experimental research has expanded to determine the long-term consequences of developmental hypoxia on adult health. Investigation has focussed on the effects of chronic prenatal hypoxia in programming cardiac dysfunction, alterations in peripheral vascular reactivity and indices of the metabolic syndrome. Several studies in rats have now reported cardiac dysfunction and an increased susceptibility to an episode of ischaemia and reperfusion (I/R) injury in hearts isolated from adult offspring of hypoxic pregnancy, particularly in males and in those fed an obesogenic diet postnatally.Reference Li, Xiao and Estrella86Reference Giussani, Camm and Niu99 Zhang and colleagues, in an elegant series of investigations, linked the reduced expression of cardio-protective genes, such as protein kinase C epsilon (PKCε) with programming an increased cardiac susceptibility to I/R injury in male offspring, as not only was the expression of PKCε reduced in hearts of hypoxic offspring, but treatment of hearts from adult offspring of normoxic pregnancy with a PKCε translocation inhibitor mimicked the defects in hearts of offspring from hypoxic pregnancy.Reference Li, Xiao and Estrella86Reference Xue, Dasgupta, Chen and Zhang91 A later study by the same group demonstrated that the mechanism via which hypoxic pregnancy caused heightened offspring cardiac susceptibility to I/R injury was epigenetic, reporting both an increase in the promoter methylation and the reduced expression of the PKCε gene in fetal pup hearts of hypoxic pregnancy, and the prevention of both effects by treatment with a DNA methylation inhibitor.Reference Patterson, Xiao, Xiong, Dixon and Zhang92 In addition to an increased risk of I/R injury, diastolic dysfunction and sympathetic dominance appear to be variables common to this cardiac phenotype in offspring of hypoxic pregnancy. Davidge and colleagues also demonstrated that adult offspring of hypoxic pregnancy have several cardiac structural and functional changes including increased expression of collagen type I and III and altered β/α myosin heavy chains ratioReference Xu, Williams, O'Brien and Davidge93 along with in vivo evidence of elevated left ventricular end diastolic pressure (LVEDP).Reference Rueda-Clausen, Morton and Davidge95 Giussani et al.Reference Giussani, Camm and Niu99 additionally reported reciprocal changes in β1-adrenergic and muscarinic receptor responsiveness in hearts from rat adult offspring of hypoxic pregnancy. Both effects are of further clinical relevance, as elevated LVEDP is associated with increased mortality,Reference Salem, Denault and Couture100 and sustained increases in myocardial contractility due to heightened sympathetic excitation and diminished parasympathetic reactivity have been strongly associated with cardiovascular disease and eventual heart failure in humans.Reference Danson, Li, Wang, Dawson and Paterson101, Reference Bristow102 Accordingly, exposure of chick embryos, mice and rat pups to hypoxia from the beginning of incubation/pregnancy promotes dilated cardiomyopathy with evidence of pump dysfunction in the offspring that persists into adulthood.Reference Sharma, Lucitti and Nordman47, Reference Tintu, Rouwet and Verlohren50, Reference Hauton98, Reference Ream, Ray, Chandra and Chikaraishi103

A significant number of studies by various groups in different species have also now reported that the adverse effects of developmental hypoxia on the offspring peripheral vasculature persist into adulthood, expressing themselves as endothelial dysfunction and the emergence of a peripheral vasoconstrictor phenotype. Ruijtenbeek et al. Reference Ruijtenbeek, Kessels and Janssen104 first reported that isolated femoral arteries of adult chickens following hypoxic incubation were more sensitive to electrical stimulation and pharmacological stimulation of peri-arterial sympathetic nerves, while showing reduced NO-dependent vasorelaxation. The developmental programming of NO-dependent endothelial dysfunction in peripheral resistance circulations has now been confirmed in adult offspring of mammalian species by the groups of Davidge and Giussani.Reference Williams, Hemmings, Mitchell, McMillen and Davidge72, Reference Hemmings, Williams and Davidge73, Reference Giussani, Camm and Niu99, Reference Morton, Rueda-Clausen and Davidge105, Reference Morton, Rueda-Clausen and Davidge106 Interestingly, two reports have shown a significant inverse relationship between low birth weight and endothelial dysfunction in children in the first decade of life and in early adulthood.Reference Leeson, Whincup and Cook107, Reference Leeson, Kattenhorn, Morley, Lucas and Deanfield108 In contrast to effects on cardiac and vascular function, studies addressing the developmental programming of indices of the metabolic syndrome by prenatal hypoxia have been restricted to three reports. Camm et al. Reference Camm, Martin-Gronert and Wright109 provided molecular evidence linking developmental hypoxia to impaired hepatic and muscle insulin signalling in adult rats, suggesting that Akt may represent a pharmaceutical target for clinical intervention against the developmental programming of metabolic disease resulting from prenatal hypoxia. The Davidge and Dyck laboratoriesReference Rueda-Clausen, Dolinsky and Morton110, Reference Dolinsky, Rueda-Clausen, Morton, Davidge and Dyck111 reported that when combined with a postnatal obesogenic diet, adult offspring of hypoxic pregnancy showed a relative increase in intra-abdominal fat deposition and adipocyte size, an increase in fasting plasma concentrations of leptin, triglyceride and free fatty acids, and an increased concentration of triglycerides and ceramides in both liver and skeletal muscle. These changes in lipid homeostasis were accompanied by in vivo insulin resistance and impaired glucose tolerance, effects that were also associated with decreased activation of Akt in the liver and skeletal muscle in response to insulin.

Intervention against programming of cardiovascular disease by prenatal hypoxia

The mechanisms via which developmental hypoxia programmes cardiovascular and metabolic diseases remain uncertain, slowing the development of clinical therapy. Several groups have raised the hypothesis that programming of cardiovascular disease by adverse developmental conditions may be secondary to oxidative stress.Reference Patterson, Xiao, Xiong, Dixon and Zhang92, Reference Giussani, Camm and Niu99, Reference Nuyt112Reference Thompson and Al-Hasan114 Giussani et al. Reference Giussani, Camm and Niu99 tested this hypothesis in relation to developmental programming by prenatal hypoxia with the first interventional study using antioxidants. The work reported that chronic prenatal hypoxia, leading to a significant increase in fetal haematocrit, promoted fetal aortic wall thickening and oxidative stress in the fetal heart and vasculature by the end of gestation. By adulthood, these effects resolved but prenatal chronic hypoxia set a functional deficit in both the heart and the peripheral circulation. Maternal treatment with vitamin C during pregnancy prevented the adverse effects in fetal offspring and reversed the enhanced myocardial contractility due to sympathetic dominance and the NO-dependent endothelial dysfunction in peripheral resistance vessels in adult offspringReference Giussani, Camm and Niu99 (Fig. 1). In that study, it was of additional interest to note that the balance of redox modulation of vascular tone, imposed by the O2:NO ratio, may be tipped in either direction to promote disequilibrium, as maternal treatment with vitamin C in normoxic pregnancy also promoted endothelial dysfunctionReference Giussani, Camm and Niu99 (Fig. 1e). Maternal antioxidant supplementation may therefore only restore the offspring vascular dysfunction in pregnancy conditions associated with increased O2 generation and vascular oxidative stress, such as during chronic prenatal hypoxia. Conversely, antioxidant treatment in healthy conditions where the offspring vascular physiology is already replenished with an appropriate redox balance may, in fact, lead to excess NO bioavailability, tipping the balance in the opposite direction. Excess NO bioavailability is known to promote peroxynitrite generation, thereby triggering mechanistic side effects resembling those of vascular oxidative stress.Reference Halliwell and Gutteridge115 The implications of these data are that maternal treatment with antioxidants may provide possible therapy against the programming effects on vascular dysfunction in complicated pregnancy, however, they clearly show that excessive vitamin C supplementation in healthy pregnancy is potentially dangerous. Patterson et al.Reference Patterson, Xiao, Xiong, Dixon and Zhang92 confirmed a role for prenatal hypoxia-derived oxidative stress in programming cardiac dysfunction, reporting that maternal treatment in rats with another antioxidant, N-acetyl-cysteine, inhibited the hypoxia-induced increase in methylation of the SP1-binding sites, reversed the decreased SP1 binding to the PKCε promoter, restored PKCε mRNA and protein abundance and abrogated the hypoxia-induced increase in susceptibility of the heart to ischaemic injury in adult offspring. Recently, they have further reported that noradrenaline causes the epigenetic repression of the PKCε gene in rodent hearts by activating Nox1-dependent ROS production.Reference Xiong, Xiao and Zhang116 Therefore, it is possible that programming of developmental hypoxia of a sympathetic dominant cardiac phenotype is mediated by catecholamine-induced ROS, which in turn causes the epigenetic repression of cardio-protective genes, such as PKCε, thereby enhancing future cardiac susceptibility to I/R injury in adult offspring. Accordingly, treatment of fetal hearts isolated from hypoxic pregnancy with a selective PKCε activator peptide ψ-εRACK, markedly improved their recovery from I/R injury.Reference Patterson, Chen, Xue, Xiao and Zhang89 Hashimoto et al.Reference Hashimoto, Pinkas and Evans117 have reported that treatment with N-acetyl cysteine of pregnant guinea pigs also inhibited the adverse effects on the fetal liver of chronic prenatal hypoxia.

Fig. 1 Cardiovascular effects of chronic developmental hypoxia with and without maternal antioxidant treatment on fetal and adult offspring. Effects on fetuses at the end of gestation (top row): (a) individual examples of fetal aortic sections; (b) mean + s.e.m. of the fetal aortic wall-to-lumen area; (c) fetal aortic nitrotyrosine staining, index of peroxinitrite generation; (d) fetal cardiac expression of HSP70, index of cardiac stress. Effects on adult offspring at 4 months of age (bottom row): (e) dilator response to methacholine expressed as overall area under the curve (AUC) in isolated femoral arteries using in vitro wire myography. Black histogram represents the AUC of the NO component and white histograms of the NO-independent component; (f) myocardial contractility of isolated heart in Langendorff preparation; (g and h) chronotropic responsiveness to muscarinic and β1-agonists, respectively. Groups are all n = 8 for normoxia (N), hypoxia (H), hypoxia with vitamin C (HC) and normoxia with vitamin C (NC). * v. N or all; †H v. HC (P < 0.05, ANOVA). Modified from Giussani et al. Reference Giussani, Camm and Niu99

It is becoming clear that enhanced oxidative stress during adverse intrauterine conditions may have a significant impact on circulations, which are highly dependent on NO, such as the utero-placental vascular bed, promoting an increase in placental vascular resistance, thereby leading to slowing of fetal growth and a reduction in birth weight. In support of this idea, it has been shown in ovine and rodent species that maternal treatment with antioxidants enhances viability and birth weight in hypoxic pregnancy.Reference Parraguez, Atlagich and Araneda118Reference Bourque, Dolinsky, Dyck and Davidge120 Two other studies have shown that maternal supplementation with melatonin protects against IUGR in a rodent and ovine model of undernourished pregnancy.Reference Richter, Hansell, Raut and Giussani121, Reference Lemley, Meyer and Camacho122 Stanley et al. Reference Stanley, Andersson and Poudel123, Reference Stanley, Andersson and Hirt124 have further reported that maternal treatment with sildenafil or tempol can rescue pup growth and improve abnormal uterine and umbilical Doppler waveforms in different knockout mice models of IUGR. The protective effects of antioxidants on fetal growth in adverse pregnancy are likely secondary to replenished NO bioavailability and improved NO-mediated umbilical perfusion, as treatment of chronically instrumented late gestation fetal sheep with either melatonin or vitamin C produced a significant increase in umbilical blood flow and vascular conductance and in vivo blockade of NO prevented the vasodilator effect.Reference Thakor, Herrera, Serón-Ferré and Giussani31

Rescue against programming of cardiovascular disease by prenatal hypoxia

While converging evidence from several laboratories points to an important role of oxidative stress and decreased NO bioavailability during hypoxic pregnancy in promoting fetal growth restriction and programming cardiovascular and metabolic dysfunction in adult life, diagnosis of fetal hypoxia and antenatal treatment with an appropriate antioxidant in a timely efficient manner may prove difficult. Therefore, the focus of research has shifted to considering how to rescue the low birth weight infant. Importantly, therapeutics for individuals diagnosed with IUGR are completely lacking. Early intervention is a potential opportunity to prevent the future development of metabolic and cardiovascular diseases, however, ethical issues exist with interventions in paediatric populations. One approach that has been raised is an intervention using Resveratrol (Resv, 97, 111), which is a natural polyphenolic antioxidant produced by plants in response to environmental stress and has demonstrated protective effects against stress and disease.Reference Baur and Sinclair125 Although there are multiple mechanisms of action for Resv, it is known to activate AMP-activated protein kinase (AMPK) as well as having antioxidant properties. AMPK is a protein kinase pathway that is involved in the control of oxidative metabolism and lipid homeostasis as well as decreasing fatty acid and triacylglycerol synthesis. Activation of AMPK in skeletal muscle also improves glucose uptake independent of insulin. Resveratrol has been shown to protect against the development of diet-induced insulin resistance in aged rodents.Reference Baur, Pearson and Price126 The Davidge and Dyck laboratoriesReference Dolinsky, Rueda-Clausen, Morton, Davidge and Dyck111 have demonstrated that early postnatal administration of Resv in the diet of weanling rats prevented features of the metabolic syndrome that are observed in offspring born from hypoxic pregnancy and fed a high fat diet. In addition, Resv prevented the diet-induced increase in plasma lipids and reduced the abdominal fat mass (Fig. 2). Moreover, Resv reduced the susceptibility to diet-induced metabolic alterations in glucose disposal and insulin resistance. The data suggest that this may be attributed to insulin sensitization of the peripheral tissues through the prevention of impaired Akt signalling as well as by activation of AMPK, thereby improving glucose utilization via an insulin-independent mechanism. In addition, postnatal administration of a fat-rich diet has been shown to be particularly deleterious to cardiac function of offspring exposed to a prenatal hypoxic insult; Resv reduced this enhanced cardiac susceptibilityReference Rueda-Clausen, Morton, Dolinsky, Dyck and Davidge97 (Fig. 2). Thus, intervention in a vulnerable paediatric patient population may reduce the risk to developing an adverse metabolic phenotype and prevent long-term cardiovascular susceptibility to disease. Additional studies are necessary to address the impact of interventions (either with Resv or other approaches) on aortic alterations and the sympathetic dominant phenotype as discussed earlier in this review.

Fig. 2 Effects of postnatal resveratrol treatment in young (3-month old) offspring exposed to prenatal hypoxia and a postnatal high fat diet. Effects on intra-abdominal fat and plasma lipids (ae): (a) mean and ±s.e.m. of total intra-abdominal fat (g); (b) percent of intra-abdominal fat to total body fat; (c) plasma triacylglycerol (TG); (d) plasma free fatty acids (FFA); (e) representative images of abdominal fat deposition by CT scan 3D reconstruction. Effects on cardiac function (fg): (f) cardiac performance during aerobic (pre-ischaemic) and after 10 min of no flow ischaemia (reperfusion); (g) average maximal cardiac power developed during the reperfusion period. Groups are all n = 6–10 for control (yellow), intrauterine growth restriction (IUGR)/prenatal hypoxia (blue), High fat (HF) – 45% fat (4.73 kcal/g), resveratrol (Resv – 4 g/kg of diet). *Represents a value of P < 0.05 for the respective main effects (IUGR or Resv) using two-way ANOVA. †P < 0.05 v. controls after a Bonferroni post hoc test comparing IUGR and control offspring receiving the same diet (n = 6 per group). Modified from Rueda-Clausen et al. Reference Rueda-Clausen, Morton, Dolinsky, Dyck and Davidge97 and Dolinsky et al.Reference Dolinsky, Rueda-Clausen, Morton, Davidge and Dyck111

Conclusions and considerations

It is now overwhelmingly clear that pregnancy complicated by fetal hypoxia can programme long-term adverse consequences on the cardiovascular health of the offspring in adult life. Interestingly, in some cases, the adverse programmed cardiovascular and metabolic phenotypes may only become evident when the offspring are exposed to additional stressors such as an obesogenic diet or ageing. Other important considerations are differences in susceptibility to disease between the sexes. While some adverse phenotypes in the adult offspring in response to prenatal hypoxia are consistent between males and females, it is important to note that sexual dimorphisms in the long-term effects of prenatal hypoxia on the cardiovascular system are evident, whereby female offspring can exhibit some degree of protection.Reference Hemmings, Williams and Davidge73 Therefore, the mechanism through which developmental insults can be modulated by sex differences is an important consideration when developing therapeutic strategies. Furthermore, the projected prevalence of cardiovascular pathophysiology in the global population may be grossly underestimated. We speculate that in the female offspring population of hypoxic pregnancy, particularly of reproductive age, programmed adverse cardiovascular and metabolic phenotypes may become significantly exacerbated when challenged by the stress of their own pregnancy. Other sensitizers to the projected prevalence of cardiovascular disease clearly include the intergenerational programming of cardiovascular pathology by prenatal hypoxia. Offspring of hypoxic pregnancy may yield offspring programmed with an increased risk of cardiovascular pathology even following normoxic pregnancy.Reference Niu, Allison and Kane127 Additional insight to mechanisms to define early interventions in pregnancy complicated by fetal hypoxia will reduce the burden not only of IUGR, but also of developmental origins of cardiovascular disease, thereby having a major clinical, economic and social impact on health.

Acknowledgements

D.G. is the Professor of Cardiovascular Physiology & Medicine at the Department of Physiology Development & Neuroscience at the University of Cambridge, Professorial Fellow and Director of Studies in Medicine at Gonville & Caius College, a Lister Institute Fellow and a Royal Society Wolfson Research Merit Award Holder. S.D. is a Canada Research Chair in Women's Cardiovascular Health and Scientist of the Alberta Innovates Health Solutions.

Financial Support

D.G. is supported by The British Heart Foundation, The Biotechnology and Biological Sciences Research Council and the Isaac Newton Trust. S.D. is supported by the Canadian Institutes of Health Research (CIHR), Heart and Stroke Foundation of Canada and the Women and Children's Health Research Institute of the University of Alberta.

Statement of Interest

None.

References

1.World Health Organization (WHO). World Health Statistics, 2012. World Health Organization: Geneva, Switzerland. ISBN 978 92 4 156444 1.Google Scholar
2.European Heart Network and European Society of Cardiology. European Cardiovascular Disease Statistics, 2012. European Heart Network and European Society of Cardiology: Brussels, Belgium. ISBN 978-2-9537898-1-2.Google Scholar
3.Conference Board of Canada. The Canadian Heart Health Strategy: Risk Factors and Future Cost Implications, 2010. Report, February 2010.Google Scholar
4.Heidenreich, PA, Trogdon, JG, Khavjou, OA, et al. Forecasting the future of cardiovascular disease in the United States: a policy statement from the American Heart Association. Circulation. 2011; 123, 933944.CrossRefGoogle ScholarPubMed
5.Agarwal, A, Williams, GH, Fisher, NDL. Genetics of human hypertension. Trends Endocrin Metab (Review). 2005; 16, 127133.CrossRefGoogle ScholarPubMed
6.Barker, DJP. Mothers, Babies and Disease in Later Life, 1994. BMJ Publishing Group: London.Google Scholar
7.Gluckman, PD, Hanson, MA, Cooper, C, Thornburg, KL. Effect of in utero and early-life conditions on adult health and disease. N Engl J Med. 2008; 359, 6173.CrossRefGoogle ScholarPubMed
8.Barker, DJP, Osmond, C, Winter, PD, Margetts, B, Simmonds, SJ. Weight in infancy and death from ischaemic heart disease. Lancet. 1989; 577580.Google Scholar
9.Rich-Edwards, JW, Stampfer, MJ, Manson, JE, et al. Birth weight and risk of cardiovascular disease in a cohort of women followed up since 1976. BMJ. 1997; 315, 396400.CrossRefGoogle Scholar
10.Leon, D, Lithell, HO, Vagero, D, et al. Reduced fetal growth rate and increased risk of death from ischaemic heart disease: cohort study of 15000 Swedish men and women born 1915–29. BMJ. 1998; 317, 241245.CrossRefGoogle Scholar
11.Fall, CHD, Stein, CE, Kumaran, K, et al. Size at birth, maternal weight, and non-insulin dependent diabetes in South India. Diabet Med. 1998; 15, 220227.3.0.CO;2-O>CrossRefGoogle ScholarPubMed
12.Huang, RC, Mori, TA, Beilin, LJ. Early life programming of cardiometabolic disease in the Western Australian pregnancy cohort (Raine) study. Clin Exp Pharmacol Physiol. 2012; 39, 973978.CrossRefGoogle ScholarPubMed
13.Levitt, NS, Lambert, EV. The foetal origins of the metabolic syndrome – a South African perspective. Cardiovasc J S Afr. 2002; 13, 179180.Google ScholarPubMed
14.Silva, AA, Santos, CJ, Amigo, H, et al. Birth weight, current body mass index, and insulin sensitivity and secretion in young adults in two Latin American populations. Nutr Metab Cardiovasc Dis. 2012; 22, 533539.CrossRefGoogle ScholarPubMed
15.Roseboom, TJ, van der Meulen, JH, Ravelli, AC, et al. Blood pressure in adults after prenatal exposure to famine. J Hypertens. 1999; 17, 325330.Google Scholar
16.Eskenazi, B, Marks, AR, Catalano, R, Bruckner, T, Toniolo, PG. Low birth weight in New York City and upstate New York following the events of September 11th. Hum Reprod. 2007; 22, 30133020.CrossRefGoogle ScholarPubMed
17.Dalziel, SR, Walker, NK, Parag, V, et al. Cardiovascular risk factors after exposure to antenatal betamethasone: 30-year follow-up of a randomised controlled trial. Lancet. 2005; 365, 18561862.CrossRefGoogle ScholarPubMed
18.McMillen, IC, Robinson, JS. Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev. 2005; 85, 571633.CrossRefGoogle ScholarPubMed
19.Seckl, JR, Meaney, MJ. Glucocorticoid programming. Ann N Y Acad Sci. 2004; 1032, 6384.CrossRefGoogle ScholarPubMed
20.Koos, BJ. Adenosine A2a receptors and O2 sensing in development. Am J Physiol Regul Integr Comp Physiol. 2011; 301, R601R622.CrossRefGoogle ScholarPubMed
21.Giussani, DA, Phillips, PS, Anstee, S, Barker, DJ. Effects of altitude versus economic status on birth weight and body shape at birth. Pediatr Res. 2001; 49, 490494.CrossRefGoogle ScholarPubMed
22.Moore, LG, Charles, SM, Julian, CG. Humans at high altitude: hypoxia and fetal growth. Respir Physiol Neurobiol. 2011; 178, 181190.CrossRefGoogle ScholarPubMed
23.Cohn, HE, Sacks, EJ, Heymann, MA, Rudolph, AM. Cardiovascular responses to hypoxemia and acidemia in fetal lambs. Am J Obstet Gynecol. 1974; 120, 817824.Google Scholar
24.Boyle, D, Hirst, K, Zerbe, G, Meschia, G, Wilkening, R. Fetal hind limb oxygen consumption and blood flow during acute graded hypoxia. Pediatr Res. 1990; 28, 94100.Google Scholar
25.Giussani, DA, Spencer, JA, Moore, PJ, Bennet, L, Hanson, MA. Afferent and efferent components of the cardiovascular reflex responses to acute hypoxia in term fetal sheep. J Physiol. 1993; 461, 431449.CrossRefGoogle ScholarPubMed
26.Jones, CT, Robinson, RO. Plasma catecholamines in foetal and adult sheep. J Physiol. 1975; 248, 1533.CrossRefGoogle ScholarPubMed
27.Fletcher, AJ, Edwards, CM, Gardner, DS, Fowden, AL, Giussani, DA. Neuropeptide Y in the sheep fetus: effects of acute hypoxemia and dexamethasone during late gestation. Endocrinology. 2000; 141, 39763982.CrossRefGoogle ScholarPubMed
28.Perez, R, Espinoza, M, Riquelme, R, Parer, JT, Llanos, AJ. Arginine vasopressin mediates cardiovascular responses to hypoxemia in fetal sheep. Am J Physiol Regul Integr Comp Physiol. 1989; 256, R1011R1018.Google Scholar
29.Morrison, S, Gardner, DS, Fletcher, AJ, Bloomfield, MR, Giussani, DA. Enhanced nitric oxide activity offsets peripheral vasoconstriction during acute hypoxaemia via chemoreflex and adrenomedullary actions in the sheep fetus. The Journal of Physiology. 2003; 547, 283291.Google Scholar
30.Thakor, AS, Richter, HG, Kane, AD, et al. Redox modulation of the fetal cardiovascular defence to hypoxaemia. J Physiol. 2010; 588, 42354247.CrossRefGoogle ScholarPubMed
31.Thakor, AS, Herrera, EA, Serón-Ferré, M, Giussani, DA. Melatonin and vitamin C increase umbilical blood flow via nitric oxide-dependent mechanisms. J Pineal Res. 2010; 49, 399406.Google Scholar
32.Kane, AD, Herrera, EA, Hansell, JA, Giussani, DA. Statin treatment depresses the fetal defence to acute hypoxia via increasing nitric oxide bioavailability. J Physiol. 2012; 590(Pt 2), 323334.Google Scholar
33.Herrera, EA, Kane, AD, Hansell, JA, et al. A role for xanthine oxidase in the control of fetal cardiovascular function in late gestation sheep. J Physiol. 2012; 590(Pt 8), 18251837.CrossRefGoogle ScholarPubMed
34.Kamitomo, M, Alonso, JG, Okai, T, Longo, LD, Gilbert, RD. Effects of long-term, high-altitude hypoxemia on ovine fetal cardiac output and blood flow distribution. Am J Obstet Gynecol. 1993; 169, 701707.CrossRefGoogle ScholarPubMed
35.Richardson, BS, Bocking, AD. Metabolic and circulatory adaptations to chronic hypoxia in the fetus. Comp Biochem Physiol A MolIntegr Physiol. 1998; 119, 717723.Google Scholar
36.Morrison, J. Sheep models of intrauterine growth restriction: fetal adaptations and consequences. Clin Exp Pharmacol Physiol. 2008; 35, 730743.Google Scholar
37.McMillen, IC, Adams, MB, Ross, JT, Coulter, CL, et al. Fetal growth restriction: adaptations and consequences. Reproduction. 2001; 122, 195204.Google Scholar
38.Bertram, CE, Hanson, MA. Animal models and programming of metabolic syndrome. Br Med Bull. 2001; 60, 103121.Google Scholar
39.Louey, S, Cock, ML, Harding, R. Postnatal development of arterial pressure: influence of the intrauterine environment. Arch Physiol Biochem. 2003; 111, 5360.Google Scholar
40.Armitage, JA, Khan, IY, Taylor, PD, Nathanielsz, PW, Poston, L. Developmental programming of the metabolic syndrome by maternal nutritional imbalance: how strong is the evidence from experimental models in mammals? J Physiol. 2004; 561, 355377.Google Scholar
41.Langley-Evans, SC, Bellinger, L, McMullen, S. Animal models of programming: early life influences on appetite and feeding behaviour. Matern Child Nutr. 2005; 1, 142148.CrossRefGoogle ScholarPubMed
42.Ruijtenbeek, K, le Noble, FAC, Janssen, GMJ, et al. Chronic hypoxia stimulates periarterial sympathetic nerve development in chicken embryo. Circulation. 2000; 102, 28922897.CrossRefGoogle ScholarPubMed
43.Miller, SL, Green, LR, Peebles, DM, Hanson, MA, Blanco, CE. Effects of chronic hypoxia and protein malnutrition on growth in the developing chick. Am J Obstet Gynecol. 2002; 186, 261267.CrossRefGoogle ScholarPubMed
44.Rouwet, EV, Tintu, AN, Schellings, MW, et al. Hypoxia induces aortic hypertrophic growth, left ventricular dysfunction, and sympathetic hyperinnervation of peripheral arteries in the chick embryo. Circulation. 2002; 105, 27912796.CrossRefGoogle ScholarPubMed
45.Ruijtenbeek, K, Kessels, LC, De Mey, JG, Blanco, CE. Chronic moderate hypoxia and protein malnutrition both induce growth retardation, but have distinct effects on arterial endothelium-dependent reactivity in the chicken embryo. Pediatr Res. 2003; 53, 573579.Google Scholar
46.Villamor, E, Kessels, CG, Ruijtenbeek, K, et al. Chronic in ovo hypoxia decreases pulmonary arterial contractile reactivity and induces biventricular cardiac enlargement in the chicken embryo. Am J Physiol Regul Integr Comp Physiol. 2004; 287, R642R651.CrossRefGoogle ScholarPubMed
47.Sharma, SK, Lucitti, JL, Nordman, C, et al. Impact of hypoxia on early chick embryo growth and cardiovascular function. Pediatr Res. 2006; 59, 116120.CrossRefGoogle ScholarPubMed
48.Tintu, AN, Noble, FA, Rouwet, EV. Hypoxia disturbs fetal hemodynamics and growth. Endothelium. 2007; 14, 353360.Google Scholar
49.Giussani, DA, Salinas, CE, Villena, M, Blanco, CE. The role of oxygen in prenatal growth: studies in the chick embryo. J Physiol. 2007; 585(Pt 3), 911917.CrossRefGoogle ScholarPubMed
50.Tintu, A, Rouwet, E, Verlohren, S, et al. Hypoxia induces dilated cardiomyopathy in the chick embryo: mechanism, intervention, and long-term consequences. PLoS One. 2009; 4, e5155.Google Scholar
51.Lindgren, I, Altimiras, J. Chronic prenatal hypoxia sensitizes beta-adrenoceptors in the embryonic heart but causes postnatal desensitization. Am J Physiol Regul Integr Comp Physiol. 2009; 297, R258R264.CrossRefGoogle ScholarPubMed
52.Salinas, CE, Blanco, CE, Villena, M, et al. Cardiac and vascular disease prior to hatching in chick embryos incubated at high altitude. J DOHaD. 2010; 1, 6066.Google Scholar
53.Lindgren, I, Altimiras, J. Sensitivity of organ growth to chronically low oxygen levels during incubation in Red Junglefowl and domesticated chicken breeds. Poult Sci. 2011; 90, 126135.Google Scholar
54.Jacobs, R, Robinson, JS, Owens, JA, Falconer, J, Webster, ME. The effect of prolonged hypobaric hypoxia on growth of fetal sheep. J Dev Physiol. 1988; 10, 97112.Google Scholar
55.Thompson, JA, Richardson, BS, Gagnon, R, Regnault, TR. Chronic intrauterine hypoxia interferes with aortic development in the late gestation ovine fetus. J Physiol. 2011; 589(Pt 13), 33193332.Google Scholar
56.Alonso, JG, Okai, T, Longo, LD, Gilbert, RD. Cardiac function during long-term hypoxemia in fetal sheep. Am J Physiol. 1989; 257(Pt 2), H581H589.Google Scholar
57.Kamitomo, M, Longo, LD, Gilbert, RD. Right and left ventricular function in fetal sheep exposed to long-term high-altitude hypoxemia. Am J Physiol. 1992; 262(Pt 2), H399H405.Google Scholar
58.Kamitomo, M, Longo, LD, Gilbert, RD. Cardiac function in fetal sheep during two weeks of hypoxemia. Am J Physiol. 1994; 266(Pt 2), R1778R1785.Google ScholarPubMed
59.Browne, VA, Stiffel, VM, Pearce, WJ, Longo, LD, Gilbert, RD. Activator calcium and myocardial contractility in fetal sheep exposed to long-term high-altitude hypoxia. Am J Physiol. 1997; 272(Pt 2), H1196H1204.Google Scholar
60.Browne, VA, Stiffel, VM, Pearce, WJ, Longo, LD, Gilbert, RD. Cardiac beta-adrenergic receptor function in fetal sheep exposed to long-term high-altitude hypoxemia. Am J Physiol. 1997; 273(Pt 2), R2022R2031.Google Scholar
61.Gilbert, RD. Fetal myocardial responses to long-term hypoxemia. Comp Biochem Physiol A Mol Integr Physiol. 1998; 119, 669674.Google Scholar
62.Kamitomo, M, Onishi, J, Gutierrez, I, Stiffel, VM, Gilbert, RD. Effects of long-term hypoxia and development on cardiac contractile proteins in fetal and adult sheep. J Soc Gynecol Investig. 2002; 9, 335341.CrossRefGoogle ScholarPubMed
63.Onishi, J, Browne, VA, Kono, S, Stiffel, VM, Gilbert, RD. Effects of long-term high-altitude hypoxia and troponin I phosphorylation on cardiac myofilament calcium responses in fetal and nonpregnant sheep. J Soc Gynecol Investig. 2004; 11, 18.Google Scholar
64.Gilbert, RD, Pearce, WJ, Longo, LD. Fetal cardiac and cerebrovascular acclimatization responses to high altitude, long-term hypoxia. High Alt Med Biol. 2003; 4, 203213.Google Scholar
65.Kim, YH, Veille, JC, Cho, MK, et al. Chronic hypoxia alters vasoconstrictive responses of femoral artery in the fetal sheep. J Korean Med Sci. 2005; 20, 1319.Google Scholar
66.Camm, EJ, Hansell, JA, Kane, AD, et al. Partial contributions of developmental hypoxia and undernutrition to prenatal alterations in somatic growth and cardiovascular structure and function. Am J Obstet Gynecol. 2010; 203, 495, e24–34.CrossRefGoogle ScholarPubMed
67.Herrera, EA, Camm, EJ, Cross, CM, et al. Morphological and functional alterations in the aorta of the chronically hypoxic fetal rat. J Vasc Res. 2012; 49, 5058.Google Scholar
68.Thompson, LP, Weiner, CP. Effects of acute and chronic hypoxia on nitric oxide-mediated relaxation of fetal guinea pig arteries. Am J Obstet Gynecol. 1999; 181, 105111.CrossRefGoogle ScholarPubMed
69.Thompson, LP, Aguan, K, Pinkas, G, Weiner, CP. Chronic hypoxia increases the NO contribution of acetylcholine vasodilation of the fetal guinea pig heart. Am J Physiol Regul Integr Comp Physiol. 2000; 279, R1813R1820.CrossRefGoogle ScholarPubMed
70.Thompson, LP. Effects of chronic hypoxia on fetal coronary responses. High Alt Med Biol. 2003; 4, 215224.Google Scholar
71.Williams, SJ, Campbell, ME, McMillen, IC, Davidge, ST. Differential effects of maternal hypoxia or nutrient restriction on carotid and femoral vascular function in neonatal rats. Am J Physiol Regul Integr Comp Physiol. 2005; 288, R360R367.Google Scholar
72.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(Pt 1), 125135.CrossRefGoogle ScholarPubMed
73.Hemmings, DG, Williams, SJ, Davidge, ST. Increased myogenic tone in 7-month-old adult male but not female offspring from rat dams exposed to hypoxia during pregnancy. Am J Physiol Heart Circ Physiol. 2005; 289, H674H682.Google Scholar
74.McEniery, CM, Wilkinson, IB. Large artery stiffness and inflammation. J Hum Hypertens. 2005; 19, 507509.CrossRefGoogle ScholarPubMed
75.Arnett, DK, Evans, GW, Riley, WA. Arterial stiffness: a new cardiovascular risk factor? Am J Epidemiol. 1994; 140, 669682.Google Scholar
76.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, e1–9.Google Scholar
77.Cruickshank, K, Riste, L, Anderson, SG, et al. Aortic pulse-wave velocity and its relationship to mortality in diabetes and glucose intolerance: an integrated index of vascular function? Circulation. 2002; 106, 20852090.CrossRefGoogle ScholarPubMed
78.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
79.Koklu, E, Kurtoglu, S, Akcakus, M, et al. Increased aortic intima-media thickness is related to lipid profile in newborns with intrauterine growth restriction. Horm Res. 2006; 65, 269275.Google Scholar
80.Akira, M, Yoshiyuki, S. Placental circulation, fetal growth, and stiffness of the abdominal aorta in newborn infants. J Pediatr. 2006; 148, 4953.Google Scholar
81.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
82.Veille, JC, Hanson, R, Sivakoff, M, Hoen, H, Ben-Ami, M. Fetal cardiac size in normal, intrauterine growth retarded, and diabetic pregnancies. Am J Perinatol. 1993; 10, 275279.CrossRefGoogle ScholarPubMed
83.Mayhew, TM, Gregson, C, Fagan, DG. Ventricular myocardium in control and growth-retarded human fetuses: growth in different tissue compartments and variation with fetal weight, gestational age, and ventricle size. Hum Pathol. 1999; 30, 655660.Google Scholar
84.Rizzo, G, Capponi, A, Rinaldo, D, Arduini, D, Romanini, C. Ventricular ejection force in growth-retarded fetuses. Ultrasound Obstet Gynecol. 1995; 5, 247255.Google Scholar
85.Miyague, NI, Ghidini, A, Fromberg, R, Miyague, LL. Alterations in ventricular filling in small-for-gestational-age fetuses. Fetal Diagn Ther. 1997; 12, 332335.CrossRefGoogle ScholarPubMed
86.Li, G, Xiao, Y, Estrella, JL, et al. Effect of fetal hypoxia on heart susceptibility to ischemia and reperfusion injury in the adult rat. J Soc Gynecol Investig. 2003; 10, 265274.Google Scholar
87.Li, G, Bae, S, Zhang, L. Effect of prenatal hypoxia on heat stress-mediated cardioprotection in adult rat heart. Am J Physiol Heart Circ Physiol. 2004; 286, H1712H1719.Google Scholar
88.Xue, Q, Zhang, L. Prenatal hypoxia causes a sex-dependent increase in heart susceptibility to ischemia and reperfusion injury in adult male offspring: role of protein kinase C epsilon. J Pharmacol Exp Ther. 2009; 330, 624632.Google Scholar
89.Patterson, AJ, Chen, M, Xue, Q, Xiao, D, Zhang, L. Chronic prenatal hypoxia induces epigenetic programming of PKC{epsilon} gene repression in rat hearts. Circ Res. 2010; 107, 365373.Google Scholar
90.Patterson, AJ, Zhang, L. Hypoxia and fetal heart development. Curr Mol Med. 2010; 10, 653666.Google Scholar
91.Xue, Q, Dasgupta, C, Chen, M, Zhang, L. Foetal hypoxia increases cardiac AT(2)R expression and subsequent vulnerability to adult ischaemic injury. Cardiovasc Res. 2011; 89, 300308.Google Scholar
92.Patterson, AJ, Xiao, D, Xiong, F, Dixon, B, Zhang, L. Hypoxia-derived oxidative stress mediates epigenetic repression of PKCε gene in foetal rat hearts. Cardiovasc Res. 2012; 93, 302310.Google Scholar
93.Xu, Y, Williams, SJ, O'Brien, D, Davidge, ST. Hypoxia or nutrient restriction during pregnancy in rats leads to progressive cardiac remodeling and impairs postischemic recovery in adult male offspring. FASEB J. 2006; 20, 12511253.Google Scholar
94.Hauton, D, Ousley, V. Prenatal hypoxia induces increased cardiac contractility on a background of decreased capillary density. BMC Cardiovasc Disord. 2009; 9, 1.Google Scholar
95.Rueda-Clausen, CF, Morton, JS, Davidge, ST. Effects of hypoxia-induced intrauterine growth restriction on cardiopulmonary structure and function during adulthood. Cardiovasc Res. 2009; 81, 713722.Google Scholar
96.Rueda-Clausen, CF, Morton, JS, Lopaschuk, GD, Davidge, ST. Long-term effects of intrauterine growth restriction on cardiac metabolism and susceptibility to ischaemia/reperfusion. Cardiovasc Res. 2011; 90, 285294.CrossRefGoogle ScholarPubMed
97.Rueda-Clausen, CF, Morton, JS, Dolinsky, VW, Dyck, JR, Davidge, ST. Synergistic effects of prenatal hypoxia and postnatal high-fat diet in the development of cardiovascular pathology in young rats. Am J Physiol Regul Integr Comp Physiol. 2012; 303, R418R426.Google Scholar
98.Hauton, D. Hypoxia in early pregnancy induces cardiac dysfunction in adult offspring of Rattus norvegicus, a non-hypoxia-adapted species. Comp Biochem Physiol A Mol Integr Physiol. 2012; 163, 278285.Google Scholar
99.Giussani, DA, Camm, EJ, Niu, Y, et al. Developmental programming of cardiovascular dysfunction by prenatal hypoxia and oxidative stress. PLoS One. 2012; 7, e31017.Google Scholar
100.Salem, R, Denault, AY, Couture, P, et al. Left ventricular end-diastolic pressure is a predictor of mortality in cardiac surgery independently of left ventricular ejection fraction. Br J Anaesth. 2006; 97, 292297.Google Scholar
101.Danson, EJ, Li, D, Wang, L, Dawson, TA, Paterson, DJ. Targeting cardiac sympatho-vagal imbalance using gene transfer of nitric oxide synthase. J Mol Cell Cardiol. 2009; 46, 482489.CrossRefGoogle ScholarPubMed
102.Bristow, MR. Beta-adrenergic receptor blockade in chronic heart failure. Circulation. 2002; 101, 558569.Google Scholar
103.Ream, M, Ray, AM, Chandra, R, Chikaraishi, DM. Early fetal hypoxia leads to growth restriction and myocardial thinning. Am J Physiol Regul Integr Comp Physiol. 2008; 295, R583R595.Google Scholar
104.Ruijtenbeek, K, Kessels, CG, Janssen, BJ, et al. Chronic moderate hypoxia during in ovo development alters arterial reactivity in chickens. Pflugers Arch. 2003; 447, 158167. 1.Google Scholar
105.Morton, JS, Rueda-Clausen, CF, Davidge, ST. Mechanisms of endothelium-dependent vasodilation in male and female, young and aged offspring born growth restricted. Am J Physiol Regul Integr Comp Physiol. 2010; 298, R930R938.Google Scholar
106.Morton, JS, Rueda-Clausen, CF, Davidge, ST. Flow-mediated vasodilation is impaired in adult rat offspring exposed to prenatal hypoxia. J Appl Physiol. 2011; 110, 10731082.Google Scholar
107.Leeson, CP, Whincup, PH, Cook, DG, 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
108.Leeson, CP, Kattenhorn, M, Morley, R, Lucas, A, Deanfield, JE. Impact of low birth weight and cardiovascular risk factors on endothelial function in early adult life. Circulation. 2001; 103, 12641268.Google Scholar
109.Camm, EJ, Martin-Gronert, MS, Wright, NL, et al. Prenatal hypoxia independent of undernutrition promotes molecular markers of insulin resistance in adult offspring. FASEB J. 2011; 25, 420427.Google Scholar
110.Rueda-Clausen, CF, Dolinsky, VW, Morton, JS, et al. Hypoxia-induced intrauterine growth restriction increases the susceptibility of rats to high-fat diet-induced metabolic syndrome. Diabetes. 2011; 60, 507516.Google Scholar
111.Dolinsky, VW, Rueda-Clausen, CF, Morton, JS, Davidge, ST, Dyck, JRB. Continued postnatal administration of resveratrol prevents diet-induced metabolic syndrome in offspring born growth restricted. Diabetes. 2011; 60, 22742284.Google Scholar
112.Nuyt, AM. Mechanisms underlying developmental programming of elevated blood pressure and vascular dysfunction: evidence from human studies and experimental animal models. Clin Sci (Lond). 2008; 114, 117.Google Scholar
113.Davidge, ST, Morton, JS, Rueda-Clausen, CF. Oxygen and perinatal origins of adulthood diseases: is oxidative stress the unifying element? Hypertension. 2008; 52, 808810.Google Scholar
114.Thompson, LP, Al-Hasan, Y. Impact of oxidative stress in fetal programming. J Pregnancy. 2012; 2012, 582748.Google Scholar
115.Halliwell, B, Gutteridge, JMC. Free Radicals in Biology and Medicine, 2004. Oxford University Press: Oxford, UK.Google Scholar
116.Xiong, F, Xiao, D, Zhang, L. Norepinephrine causes epigenetic repression of PKCε gene in rodent hearts by activating Nox1-dependent reactive oxygen species production. FASEB J. 2012; 26, 27532763.Google Scholar
117.Hashimoto, K, Pinkas, G, Evans, L, et al. Protective effect of N-acetylcysteine on liver damage during chronic intrauterine hypoxia in fetal guinea pig. Reprod Sci. 2012; 19, 10011009.Google Scholar
118.Parraguez, VH, Atlagich, M, Araneda, O, et al. Effects of antioxidant vitamins on newborn and placental traits in gestations at high altitude: comparative study in high and low altitude native sheep. Reprod Fertil Dev. 2011; 23, 285296.CrossRefGoogle ScholarPubMed
119.Richter, HG, Camm, EJ, Modi, BN, et al. Ascorbate prevents placental oxidative stress and enhances birth weight in hypoxic pregnancy in rats. J Physiol. 2012; 590(Pt 6), 13771387.Google Scholar
120.Bourque, SL, Dolinsky, VW, Dyck, JR, Davidge, ST. Maternal resveratrol treatment during pregnancy improves adverse fetal outcomes in a rat model of severe hypoxia. Placenta. 2012; 33, 449452.Google Scholar
121.Richter, HG, Hansell, JA, Raut, S, Giussani, DA. Melatonin improves placental efficiency and birth weight and increases the placental expression of antioxidant enzymes in undernourished pregnancy. J Pineal Res. 2009; 46, 357364.Google Scholar
122.Lemley, CO, Meyer, AM, Camacho, LE, et al. Melatonin supplementation alters uteroplacental hemodynamics and fetal development in an ovine model of intrauterine growth restriction. Am J Physiol Regul Integr Comp Physiol. 2012; 302, R454R467.Google Scholar
123.Stanley, JL, Andersson, IJ, Poudel, R, et al. Sildenafil citrate rescues fetal growth in the catechol-O-methyl transferase knockout mouse model. Hypertension. 2012; 59, 10211028.Google Scholar
124.Stanley, JL, Andersson, IJ, Hirt, CJ, et al. Effect of the anti-oxidant tempol on fetal growth in a mouse model of fetal growth restriction. Biol Reprod. 2012; 87, 251258.Google Scholar
125.Baur, JA, Sinclair, DA. Therapeutic potential of resveratrol: the in vivo evidence. Nature Reviews Drug Discovery. 2006; 5, 493506.Google Scholar
126.Baur, JA, Pearson, KJ, Price, NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006; 444, 337342.CrossRefGoogle ScholarPubMed
127.Niu, Y, Allison, BJ, Kane, AD, et al. Intergenerational inheritance of cardiovascular disease risk induced by chronic fetal hypoxia. Society for Gynecologic Investigation, 60th Annual Scientific Meeting, March 20–23, 2013, Orlando, FL, USA.Google Scholar
Figure 0

Fig. 1 Cardiovascular effects of chronic developmental hypoxia with and without maternal antioxidant treatment on fetal and adult offspring. Effects on fetuses at the end of gestation (top row): (a) individual examples of fetal aortic sections; (b) mean + s.e.m. of the fetal aortic wall-to-lumen area; (c) fetal aortic nitrotyrosine staining, index of peroxinitrite generation; (d) fetal cardiac expression of HSP70, index of cardiac stress. Effects on adult offspring at 4 months of age (bottom row): (e) dilator response to methacholine expressed as overall area under the curve (AUC) in isolated femoral arteries using in vitro wire myography. Black histogram represents the AUC of the NO component and white histograms of the NO-independent component; (f) myocardial contractility of isolated heart in Langendorff preparation; (g and h) chronotropic responsiveness to muscarinic and β1-agonists, respectively. Groups are all n = 8 for normoxia (N), hypoxia (H), hypoxia with vitamin C (HC) and normoxia with vitamin C (NC). * v. N or all; †H v. HC (P < 0.05, ANOVA). Modified from Giussani et al.99

Figure 1

Fig. 2 Effects of postnatal resveratrol treatment in young (3-month old) offspring exposed to prenatal hypoxia and a postnatal high fat diet. Effects on intra-abdominal fat and plasma lipids (ae): (a) mean and ±s.e.m. of total intra-abdominal fat (g); (b) percent of intra-abdominal fat to total body fat; (c) plasma triacylglycerol (TG); (d) plasma free fatty acids (FFA); (e) representative images of abdominal fat deposition by CT scan 3D reconstruction. Effects on cardiac function (fg): (f) cardiac performance during aerobic (pre-ischaemic) and after 10 min of no flow ischaemia (reperfusion); (g) average maximal cardiac power developed during the reperfusion period. Groups are all n = 6–10 for control (yellow), intrauterine growth restriction (IUGR)/prenatal hypoxia (blue), High fat (HF) – 45% fat (4.73 kcal/g), resveratrol (Resv – 4 g/kg of diet). *Represents a value of P < 0.05 for the respective main effects (IUGR or Resv) using two-way ANOVA. †P < 0.05 v. controls after a Bonferroni post hoc test comparing IUGR and control offspring receiving the same diet (n = 6 per group). Modified from Rueda-Clausen et al.97 and Dolinsky et al.111