Hostname: page-component-7c8c6479df-r7xzm Total loading time: 0 Render date: 2024-03-29T07:58:57.881Z Has data issue: false hasContentIssue false

The conflicting effects of maternal nutrient restriction and early-life obesity on renal health

Published online by Cambridge University Press:  14 January 2011

H. P. Fainberg*
Affiliation:
School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK
H. Budge
Affiliation:
Early Life Nutrition Research Unit and Respiratory Medicine Biomedical Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital, Nottingham NG7 2UH, UK
M. E. Symonds
Affiliation:
Early Life Nutrition Research Unit and Respiratory Medicine Biomedical Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital, Nottingham NG7 2UH, UK
*
*Corresponding author: Dr Hernan P Fainberg, fax: +44 115 951 6415, email Pablo.Fainberg@nottingham.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Epidemiological and animal studies have demonstrated that early-life nutrition alters the metabolic responses and generates structural changes in complex tissues, such as the kidneys, which may lead to a reduction in the offspring lifespan. Independently, obesity induces a spontaneous low-grade chronic inflammatory response by modulating several of the major metabolic pathways that ultimately compromise long-term renal health. However, the combined effects of maternal nutrition and early-life obesity in the development of renal diseases are far from conclusive. Previous results, using the ovine model, demonstrated that the combination of a reduction in fetal nutrition and juvenile obesity induced a series of adaptations associated with severe metabolic syndrome in the heart and adipose tissue. Surprisingly, exposure to an obesogenic environment in the kidney of those offspring produced an apparent reduction in glomerulosclerosis in relation to age- and weight-matched controls. However, this reduction in cellular apoptosis was accompanied by a rise in glomerular filtration rate and blood pressure of equal intensity when compared with obese controls. The intention of this review is to explain the adaptive responses observed in this model, based on insights into the mechanism of renal fetal programming, and their potential interactions with some of the metabolic changes produced by obesity.

Type
Irish Section Postgraduate Symposium
Copyright
Copyright © The Authors 2011

Abbreviations:
GH

growth hormone

IGF

insulin-like growth factor

RAS

renin–angiotensin system

Deterioration in renal health is the result of both heritable and environmental factors which, in turn, influence the rate of functional decline. A clear environmental factor that reduces kidney function is that produced with chronic inflammation and induced by metabolic disorders, such as obesity(Reference Hsu, McCulloch and Iribarren1, Reference Eriksson, Forsen and Tuomilehto2). A good example of the intricate relationship between the nutritional environment and metabolic-renal-associated diseases at the population level is observed in previously underdeveloped countries, such as China, a society that has experienced rapid growth in the prevalence of obesity due to several changes in lifestyle(Reference Wang, Kong and Wu3). In less than a generation, medical conditions associated with overweight or obesity, including type 2 diabetes, hypertension and renal diseases, have increased to unprecedented levels, matching those existing in western developed countries. Unfortunately, this rapid change in the nutritional environment has exposed an inherent human vulnerability to the complications of excess weight gain particularly in young children(Reference Wang, Kong and Wu3).

In an attempt to explain the effects of early-life nutrition observed in previous epidemiological studies and their association with the development of metabolic complications in later life, Barker and Hales proposed the ‘thrifty phenotype hypothesis’(Reference Hales and Barker4). The basis of this hypothesis is that the maternal nutritional environment and early postnatal life nutrition play a major role in the pathogenesis of renal-CVD by affecting tissue structure and function, in conjunction with other multiple mechanisms, which alter the hormonal milieu of the offspring(Reference Hales and Barker4). Today, there is good evidence supporting the influence of the composition of in utero nutrition with a predisposition to develop cardiovascular and renal complications in later life(Reference Barker, Osmond and Golding5Reference Zidar, Avgustin Cavic and Kenda7). Importantly, extensive animal studies have demonstrated that alterations in maternal diet can permanently alter renal structure and function, which, depending on the nutritional composition, may result in substantial reduction in offspring longevity, thus validating the principles of the thrifty phenotype hypothesis(Reference Ozanne and Hales8).

The relationship between renal programming and the maternal environment

During the early- and mid-20th century, at least in industrialised countries, maternal and early-life undernutrition were important factors affecting long-term health(Reference Kermack, McKendrick and McKinlay9). For that reason, Barker and other epidemiologists based their research on those groups of society that suffered chronic food shortages or imbalances in dietary composition (micro- and macronutrients) during pregnancy or early infancy. Not surprisingly, these early studies attributed radical declines in renal–vascular function to differences in birth weight(Reference Barker, Osmond and Golding5, Reference Hales, Barker and Clark10, Reference Nelson, Morgenstern and Bennett11). However, more recent analysis of renal autopsies of adults born in the lower range of birth weight showed substantial variation in renal composition, revealing the possibility of an innate ability of the kidney to overcome an adverse fetal environment(Reference Hughson, Farris and Douglas-Denton12).

The apparent natural capacity of the kidney to alleviate the negative effects of low birth weight were also observed and described in other epidemiological studies, such as those conducted on adults exposed in utero to the Dutch winter famine of 1944–45 (at the peak of which daily rations declined to under 8368 kJ/d (2000 kcal/d)).(Reference Painter, Roseboom and Bleker13) By dividing the period of malnutrition into trimesters, the authors of these investigations were able to determine very different long-term health outcomes for the adult offspring(Reference Painter, Roseboom and Bleker13). These studies demonstrated that the only group of individuals to develop a long-term tendency towards renal–vascular deterioration were those exposed to the famine around the second trimester of gestation(Reference Painter, Roseboom and Bleker13). In addition, this group of offspring, as mature adults, showed an increased risk of developing insulin resistance and other traits of the metabolic syndrome, including obesity(Reference Ravelli, van der Meulen and Michels14). The important contribution of these cohorts is to show that the time at which nutrient supply to the fetus was reduced produced specific health outcomes in later life, irrespective of birth weight, thus emphasising the importance of the different stages of embryonic and fetal development(Reference Ravelli, van der Meulen and Michels14Reference Hoek, Brown and Susser16).

Potential mechanisms linking the maternal nutritional environment and renal development

Early renal embryonic development involves extensive cell proliferation to form different complex structures on which maternal nutritional and metabolic environment may play an important role(Reference Brennan, Gopalakrishnan and Kurlak17, Reference Moritz, Boon and Wintour18). Thus, the subsequent cycle of renal cell differentiation, including proliferation as well as extensive cell death to eliminate not only damaged cells but also immature ones, is clearly a process that may have lasting and permanent consequences for the offspring(Reference Bard19). It is important to mention that the formation of the early embryonic renal structures that subsequently form the nephrons (the basic units of renal filtration) starts around the first week of gestation. In human subjects, the first permanent structures of future nephrons commence their formation at about the fifth week of gestation, and at about week 32, the first fully developed nephrons appear in the fetal kidney(Reference Brenner and Rector20). Although these structures continue to mature in the days before birth and after birth, a total inhibition of the formation of new nephrons in large mammals, such as human subjects and sheep, extends throughout their lifetime. At the time of birth, the total number of nephrons per kidney is around 1 million(Reference Brennan, Gopalakrishnan and Kurlak17, Reference Moritz, Boon and Wintour18).

All these processes that regulate the intra-uterine renal development, as expected, involve a large number of fetal genes and hormones; although the maternal hormonal milieu and the nutrient supply to the fetus influence its development(Reference Hughson, Farris and Douglas-Denton12, Reference Bard19, Reference Godfrey, Robinson and Barker21).

The following section describes some of the fetal hormonal axes known to interact with the maternal nutritional status during gestation, which may influence renal development.

The role of the insulin-like growth factor axis

In human subjects and rodents, manipulating the supply of nutrients to the fetus produces alterations in cord blood concentration of several anabolic hormones, including the different members of the insulin-like growth factor (IGF) family (IGF-I and -II), their receptors and binding proteins(Reference Ong and Dunger22). During gestation, the receptors of this hormone family are detectable in growing kidneys and other tissues, including the adipose tissue(Reference Ymer and Herington23, Reference Holzenberger, Hamard and Zaoui24). Furthermore, the crucial role of the IGF-I receptor in fetal renal development was confirmed in transgenic mouse models(Reference Ymer and Herington23Reference Hirschberg and Kopple25). The lack of activity of this receptor during renal embryonic differentiation had a number of negative effects on the renal constitution of those rodents including a reduction in the number of nephrons and other irregularities in gomerular morphology that would contribute to the progression of renal disease(Reference Hirschberg and Kopple25Reference Bridgewater, Dionne and Butt27).

The role of leptin

Leptin is another hormone associated with maternal diet that may have an impact on the developing offspring(Reference Schubring, Siebler and Kratzsch28, Reference Schubring, Kiess and Englaro29). Circulating concentrations of leptin in the mother are proportional to her fat mass. Normally, adipocytes are the primary source of leptin, which then interacts with receptors in the hypothalamus, to inhibit food intake by acting through neuropeptide Y(Reference Schwartz, Baskin and Bukowski30).

The concentration and function of leptin varies depending on the stage of gestation. For instance, during the last third of pregnancy in human subjects, the production of leptin by adipose tissue declines while there is an increased secretion of this hormone by the placenta and fetus(Reference Schubring, Kiess and Englaro29, Reference Bell and Ehrhardt31Reference Bispham, Gopalakrishnan and Dandrea33). Furthermore, a reduction of 40% of maternal food intake during early to mid-gestation (3·6 MJ/d, on days 28 to 80 (145 to full-term)) has been shown to produce a decline in leptin plasma concentrations in comparison with control-fed mothers.

Although the direct effects of leptin in the fetal kidney are unknown, the localisation of its receptors and its gene expression in fetal perirenal adipose tissue, bone and cartilage, suggest that this hormone has a role in the control of early growth, which may influence renal function in later life(Reference Bispham, Gopalakrishnan and Dandrea33). In mature kidneys, five isoforms of the leptin receptors are known to be active and these are mainly located in the inner medullar(Reference Serradeil-Le Gal, Raufaste and Brossard34). In vivo studies demonstrate that leptin can regulate Na handling and increase renal sympathetic nerve activity, which are early signs of hypertension(Reference Beltowski, Jamroz-Wisniewska and Borkowska35, Reference Hall36). Importantly, although in obese individuals there is a partial lack of function of leptin in the hypothalamus its renal actions are unaffected(Reference Rahmouni, Morgan and Morgan37).

The role of glucocorticoids and other hormones

Thyroxine and cortisol hormone concentrations, which are involved in vascular development of the newborn, are also compromised by changes in maternal nutritional intervention(Reference Moritz, Boon and Wintour18, Reference Bispham, Gopalakrishnan and Dandrea33). Bispham and colleagues observed, when using an ovine model, that changes in maternal thyroxine and leptin are accompanied by an increase in NEFA in the plasma of nutrient-restricted mothers during mid-gestation (3·5 MJ/d, on days 30 to 80), without a change in glucose concentration. The authors concluded that the increase in lipolysis may act as a physiological response to sustain the glucose supply for the optimal development of the offspring and the placenta during the nutritional challenge. However, this adaptation was insufficient to avoid a reduction in placental growth; although the subsequent weight of the offspring was similar to that of their controls(Reference Bispham, Gopalakrishnan and Dandrea33). A reduction in maternal cortisol secretion may have important consequences for the endocrine programming of the offspring, due to its effects in the maturation of the hypothalamic-pituitary adrenal axis, but these are yet to be confirmed. In turn, changes in the activation of the glucocorticoids complex may alter the control of important physiological and other endocrine functions, including adipogenesis and the renin–angiotensin system (RAS), affecting renal function(Reference Moritz, Boon and Wintour18).

Changes in maternal diet during mid-gestation in sheep and their consequences on the offspring

In an attempt to understand the biological mechanisms behind the association between maternal diet, particularly around the period of fetal renal development, and long-term physiological changes in the offspring, several animal models have been proposed. Between the different animal models studied, the ovine model, due its relatively long pregnancy (145 d) and its ability to produce an offspring of similar weight to human subjects, with fully developed organs, has proven to be useful for increasing the understanding of the effects of changes in maternal diet at specific stages of pregnancy(Reference Symonds, Stephenson and Gardner38).

In different ovine studies, it was observed that a reduction of 50% in maternal food intake up to mid-gestation (days 30 to 80 (145 d to full-term)) produced an offspring of weight similar to those born to control-fed mothers(Reference Whorwood, Firth and Budge39, Reference Williams, Kurlak and Perkins40). However, as noted in the human Dutch winter cohorts, kidney physiology, blood pressure and fat mass were affected at different points in the offspring's life(Reference Whorwood, Firth and Budge39, Reference Bispham, Gardner and Gnanalingham41, Reference Gopalakrishnan, Gardner and Rhind42). Near-term nutrient-restricted offspring (days 140 to 145 (145 d to full-term)) exhibited increase in the gene expression of a range of factors associated with the development of adipose tissue, including IGF-I and -II(Reference Bispham, Gopalakrishnan and Dandrea33). In the same model, adaptations were also observed in other key regulatory components of fat metabolism, including gene expression of the mitochondrial uncoupling protein 2 and PPARα. All these responses were accompanied by increased adiposity(Reference Bispham, Gardner and Gnanalingham41). In the newborn kidney, an increase in renal length was observed, followed by an increase in the gene expression of the receptors for glucocorticoids and angiotensin, particularly the type 1 isoform(Reference Whorwood, Firth and Budge39). Therefore, an important factor affecting the long-term health of the offspring is body composition at birth, which is indirect evidence of an unfavourable maternal metabolic and hormonal environment during earlier pregnancy(Reference Symonds, Sebert and Hyatt43).

Changes in body composition, influenced by maternal nutrition, may have long-term consequences for the offspring. In a similar maternal nutritional intervention (30–80 d of gestation), six-month-old offspring showed a reduction in nephron number, which was linked to cellular apoptosis, followed by a reduction in blood pressure, in relation to those offspring born to control-fed mothers(Reference Gopalakrishnan, Gardner and Dandrea44). One of the few long-term studies on the effects of early to mid-gestation maternal nutrient restriction (3·5 MJ/d, from 0 to 95 d of gestation) demonstrated that, at three years of age, offspring born to nutrient-restricted mothers were transiently hypertensive prior to feeding and the heart rate response to noradrenaline infusion blunted, which is a sign of cardiovascular decline. In addition, the hormonal plasma profile indicated an increase in leptin concentration that correlated with greater fat mass(Reference Gopalakrishnan, Gardner and Rhind42). These results indicated that maternal nutrient restriction during early to mid-gestation alters the long-term physiological and endocrine responses in the kidney and other tissues, including adipose tissue and could ultimately increase the risk of CVD in later life(Reference Gopalakrishnan, Gardner and Dandrea44).

Early life nutrition and its subsequent effects on renal health

In addition to the maternal nutritional environment postnatal diet may also affect later renal health. Human epidemiological and animal studies indicate that early infant nutrition, independently of the in utero environment, modulates susceptibility to chronic diseases in adulthood(Reference Stein, Fall and Kumaran45). During the first nine months of postnatal life, there is an accelerated and linear period of growth, influenced by the endocrine actions of growth hormone (GH), the secretion of which is regulated mainly by nutritional intake(Reference Eriksson, Forsen and Tuomilehto2, Reference Ogilvy-Stuart, Hands and Adcock46, Reference Low, Tam and Kwan47). The secretion of GH induces the production of IGF-I which, in turn, binds to its own receptor (IGF-I receptor) and triggers growth. However, the binding of IGF-I to IGF-I receptor inhibits the actions of GH, particularly in young animals. These actions of GH regulated by the nutritional environment persist until one year of age in human subjects(Reference Tannenbaum, Guyda and Posner48).

In addition to having a role in general physiological development, several observations indicate that the IGF–GH axis has a role in renal function. For instance, the infusion of GH, through the action of IGF-I, resulted in an increase in glomerular filtration rate and renal plasma flow(Reference Hirschberg and Kopple25). Possibly for that reason, it was observed that individuals, who, as adults, were overweight and also suffered from rapid vascular deterioration, experienced as young infants rapid increase in height and weight(Reference Eriksson, Forsen and Tuomilehto2).

Adult obesity and renal health

Independent of diet in early life, exposure to a juvenile or adult obesogenic environment also produces a significant deterioration in renal health through similar endocrine mechanisms. In human subjects and animal models, it was observed that obesity produced an increase in a series of haemodynamic changes, indicated by an elevation in blood pressure followed by initial elevation of renal plasma flow as well as glomerular filtration rate, which then with time is accompanied by variable degrees of proteinuria(Reference Hall36, Reference Adelman, Restaino and Alon49, Reference Chagnac, Weinstein and Korzets50). Some observations indicate that inflammation, in particular of the renal proximal tubules, may be one of the first morphological changes linked to kidney disease in obese non-diabetic individuals(Reference Praga, Hernandez and Herrero51, Reference Rea, Heimbach and Grande52). Inflammation is an important feature in obesity-associated diseases; several cohorts demonstrated a correlation between increased serum concentrations of pro-inflammatory molecules of obese individuals and markers of renal disease, such as microalbuminuria(Reference Nakamura, Onoda and Itai53, Reference Ramkumar, Cheung and Pappas54). In severely obese human subjects with a BMI >30 kg/m2, signs of advanced renal dysfunction, such as the appearance of ectopic lipid deposition, are accompanied by the secretion of pro-inflammatory molecules and other hormones, including angiotensin II, by the adipose tissue(Reference Hotamisligil, Shargill and Spiegelman55Reference Fried, Bunkin and Greenberg57).

The production of angiotensin II by the adipose tissue and its effects on the kidney

Angiotensin II (the active component of the RAS), in addition to acting as a key regulator of arterial blood pressure, can activate multiple cellular responses in renal and adipose tissue, which are associated with cellular proliferation and stress(Reference Massiera, Bloch-Faure and Ceiler58, Reference van Harmelen, Skurk and Rohrig59). RAS is a complex enzymatic cascade, involving several tissues and hormones activated by alterations in blood pressure, volume or simple reductions in blood electrolytes (mainly NaCl concentration) causing a range of physiological responses including contraction of vascular vessels and Na retention. In lean individuals, the majority of angiotensinogen (one hormone in this cascade) is secreted into the circulation by the liver. Later, it is transformed to angiotensin I through the catalytic action of renin, an enzyme produced by granular cells residing in the renal juxtaglomerular apparatus, and converted by an angiotensin-converting enzyme to angiotensin II(Reference Brenner and Rector20). However, adipocytes can also secrete angiotensinogen and, with obesity, this increases in parallel with raised lipid accumulation by mature adipocytes(Reference Aubert, Darimont and Safonova60).

The increase in angiotensin II concentrations has been associated with renal injury, in part mediated by its own vasoconstrictive actions, which trigger a rise in the production of reactive oxidative species(Reference Hasdan, Benchetrit and Rashid61). In the first instance, increased exposure to angiotensin II induces a rise in cell proliferation, particularly of mesangial cells(Reference Johnson, Alpers and Yoshimura62). As observed with other hormones, plasma concentrations of angiotensin II increase with obesity and, as noted for IGF-I and insulin, angiotensin II is also associated with lipid and collagen accumulation in renal tissue(Reference Abrass, Raugi and Gabourel63Reference Saito, Ishizaka and Hara65). Long-term exposure to angiotensin II, due to its contractile actions as well as the subsequent increase in metabolic activity in proximal tubules, leads to an increase in oxygen consumption producing renal ischaemia(Reference Lauzier, Page and Michaud66). The resultant reduction in oxygen supply and the rise of oxidative stress in the glomerulus and proximal tubules may be the first stages leading to cell cycle arrest or apoptosis; cellular processes that characterise several renal diseases associated with obesity(Reference Lubbers and Baumgartl67, Reference Wiggins, Goyal and Sanden68). However, epidemiological evidence has demonstrated that in obese patients this process of renal health decline may be prolonged for years(Reference Praga, Hernandez and Morales69). This may be due to the innate ability of the kidneys to adapt to low-oxygen tensions by an efficient tissue neovascularisation, which is a major difference from adipose tissue, allowing it to overcome some of the metabolic changes induced by obesity(Reference Chagnac, Weinstein and Korzets50, Reference Lauzier, Page and Michaud66, Reference Advani, Kelly and Advani70).

The combined effects of maternal nutrient restriction and obesity in juvenile sheep

In sheep, as observed in other animal models, including human subjects, an expansion of adipose tissue produces a rise in plasma leptin, NEFA, catecholamines and insulin, which was the only systemic hormone when secretion was amplified in offspring previously exposed to maternal nutrient restriction (3·5 MJ/d, from days 28 to 145 (term=147 d of gestation))(Reference Williams, Kurlak and Perkins40, Reference Sebert, Hyatt and Chan71). Surprisingly, other plasma metabolites also associated with metabolic dysfunction, such as glucose, cortisol and TAG, were unaffected by the increase in adult body weight(Reference Sebert, Hyatt and Chan71).

Despite the similarities in plasma profiles between both obese groups (i.e. obese with maternal nutritional restriction and obese with normal maternal diet), only the adipose tissue of obese sheep born to nutrient-restricted mothers showed similarities to tissue from animal models exposed to severe obesity, such as an increase in the number of necrotic adipocytes and the secretion of pro-inflammatory factors linked with macrophage infiltration(Reference Cinti, Mitchell and Barbatelli56, Reference Sharkey, Gardner and Fainberg72). These responses were accompanied by endoplasmic reticulum stress, a possible response to a reduction in oxygen supply to the adipose tissue(Reference Sharkey, Gardner and Fainberg72). In other tissues associated with the reno–cardiovascular system, these adaptations in the adipose tissue are followed by a subsequent increase in ectopic lipid deposition in the heart(Reference Chan, Sebert and Hyatt73).

From the point of view of the cardiovascular system, the increase in fat mass generated considerable structural changes in the heart, particularly in the left ventricular region, irrespective of the early-life environment(Reference Chan, Sebert and Hyatt73). In comparison with obese controls, offspring exposed to maternal nutrient restriction showed important alterations in myocardial energy metabolism, as indicated by an increase in ectopic lipid deposition and by a subsequent reduction in the expression of genes associated with lipid cellular transport and with severe metabolic dysfunction(Reference Chan, Sebert and Hyatt73, Reference Zhou, Grayburn and Karim74). However, the nutrient-restricted obese group showed a blunted cardiac parasympathetic response to atropine infusion(Reference Chan, Sebert and Hyatt73).

Changes in kidney function and structure in sheep

Obesity in sheep, as reported in human subjects, causes increases in blood pressure and glomerular filtration rate, which are important signs of renal dysfunction(Reference Chagnac, Weinstein and Korzets50). In the heart and in the kidney, significant changes occurred in gene expression between the maternal nutrient-restricted group and their obese controls, principally of factors associated with exposure to cytokines and angiotensin II, possibly suggesting an enhanced adaptation to obesity(Reference Williams, Kurlak and Perkins40). However, similar increases in haemodynamic parameters may suggest similar vasomotor afferent responses in both obese groups to the RAS, which also include extensive cell proliferation, particularly of mesangial and proximal tubular endothelial cells(Reference Hall36). This suggests that the substantial changes in the composition in the kidney of those offspring exposed to maternal nutrient restriction, potentially may include an increase in lipid deposition(Reference Chan, Sebert and Hyatt73). We have also demonstrated that the cellular apoptosis observed in the obesity group was associated with endoplasmic reticulum stress that, in the kidney, is a temporary adaptation to a reduction in oxygen supply(Reference Sharkey, Gardner and Fainberg72). However, the activation of this response may lead, in extreme circumstances, to significant renal injury(Reference Sharkey, Gardner and Fainberg72) as summarised in Fig. 1. Therefore, we conclude that the ovine model mimics many of the characteristics observed in obese human subjects and in other animal models(Reference Rea, Heimbach and Grande52, Reference Henegar, Bigler and Henegar75).

Fig. 1. Diagrammatic representation of renal responses in offspring born to mid-gestation nutrient-restricted (NR) mothers compared to their control (C) group, as observed previously in sheep. Exposure to an obesogenic environment induced a series of hormonal responses triggered by changes in adipocyte responses to excess lipid accumulation leading to tissue remodelling and alterations in renal and related peripheral tissues. RAS, renin–angiotensin system; IGF, insulin-like growth factor.

Conclusions

Human and animal studies have demonstrated the notable influence of maternal nutritional environment on the offspring growth trajectory, and its impact in the development of non-transmissible diseases in later life. The prevalence of obesity worldwide, particularly in children, makes us re-evaluate policies involving maternal and infant nutrition. As noted in this review, obesity per se, even accounting for different types of maternal diet, undoubtedly produces an extensive decline in renal function, and has a deleterious effect on various other tissues, including adipose tissue. However, by altering the in utero nutritional environment, it is possible to observe different phenotypes, which are more distinct when those individuals are exposed to an obesogenic environment.

Obesity, through metabolic disarrangement, produces many structural and functional alterations in the kidney that can differ according to the in utero experience. In order to understand the alterations induced by both maternal and early nutritional environment, it is necessary to follow up offspring over an extensive period of time, analysing development through a range of physiological and metabolic measurements. One potential link between the changes triggered by alterations on the maternal–fetal nutrition and the following metabolic derangements during obesity may include functional alterations of adipose tissue and the subsequent resetting of the RAS, which has an adverse impact on renal health.

Acknowledgements

The authors acknowledge the support of the British Heart Foundation, the European Union Sixth Framework for Research and Technical Development of the European Community – The Early Nutrition Programming Project (FOOD-CT-2005-007036) and the Nottingham Respiratory Medicine Biomedical Research Unit in their research. The authors declare no conflicts of interest. The main author of the paper was H. P. Fainberg, under the supervision of H. B. and M. E. S.

References

1.Hsu, CY, McCulloch, CE, Iribarren, C et al. (2006) Body mass index and risk for end-stage renal disease. Ann Intern Med 144, 2128.CrossRefGoogle ScholarPubMed
2.Eriksson, JG, Forsen, T, Tuomilehto, J et al. (2001) Early growth and coronary heart disease in later life: longitudinal study. Br Med J 322, 949953.CrossRefGoogle ScholarPubMed
3.Wang, L, Kong, L, Wu, F, et al. (2005) Preventing chronic diseases in China. Lancet 366, 18211824.CrossRefGoogle ScholarPubMed
4.Hales, CN & Barker, DJ (2001) The thrifty phenotype hypothesis. Br Med Bull 60, 520.CrossRefGoogle ScholarPubMed
5.Barker, DJ, Osmond, C, Golding, J et al. (1989) Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. Br Med J 298, 564567.CrossRefGoogle ScholarPubMed
6.Wadsworth, ME, Cripps, HA, Midwinter, RE et al. (1985) Blood pressure in a national birth cohort at the age of 36 related to social and familial factors, smoking, and body mass. Br Med J (Clin Res Ed) 291, 15341538.CrossRefGoogle Scholar
7.Zidar, N, Avgustin Cavic, M, Kenda, RB et al. (1998) Unfavorable course of minimal change nephrotic syndrome in children with intrauterine growth retardation. Kidney Int 54, 13201323.CrossRefGoogle ScholarPubMed
8.Ozanne, SE & Hales, CN (2004) Lifespan: catch-up growth and obesity in male mice. Nature 427, 411412.CrossRefGoogle ScholarPubMed
9.Kermack, WO, McKendrick, AG & McKinlay, PL (2001) Death-rates in Great Britain and Sweden. Some general regularities and their significance. Int J Epidemiol 30, 678683.CrossRefGoogle ScholarPubMed
10.Hales, CN, Barker, DJ, Clark, PM et al. (1991) Fetal and infant growth and impaired glucose tolerance at age 64. Br Med J 303, 10191022.CrossRefGoogle ScholarPubMed
11.Nelson, RG, Morgenstern, H & Bennett, PH (1998) Birth weight and renal disease in Pima Indians with type 2 diabetes mellitus. Am J Epidemiol 148, 650656.CrossRefGoogle ScholarPubMed
12.Hughson, M, Farris, AB 3rd, Douglas-Denton, R et al. (2003) Glomerular number and size in autopsy kidneys: the relationship to birth weight. Kidney Int 63, 21132122.CrossRefGoogle ScholarPubMed
13.Painter, RC, Roseboom, TJ & Bleker, OP (2005) Prenatal exposure to the Dutch famine and disease in later life: an overview. Reprod Toxicol 20, 345352.CrossRefGoogle Scholar
14.Ravelli, AC, van der Meulen, JH, Michels, RP et al. (1998) Glucose tolerance in adults after prenatal exposure to famine. Lancet 351, 173177.CrossRefGoogle ScholarPubMed
15.Roseboom, TJ, van der Meulen, JH, Osmond, C et al. (2000) Coronary heart disease after prenatal exposure to the Dutch famine, 1944–1945. Heart 84, 595598.CrossRefGoogle Scholar
16.Hoek, HW, Brown, AS & Susser, E (1998) The Dutch famine and schizophrenia spectrum disorders. Soc Psychiatry Psychiatr Epidemiol 33, 373379.CrossRefGoogle ScholarPubMed
17.Brennan, KA, Gopalakrishnan, GS, Kurlak, L et al. (2005) Impact of maternal undernutrition and fetal number on glucocorticoid, growth hormone and insulin-like growth factor receptor mRNA abundance in the ovine fetal kidney. Reproduction 129, 151159.CrossRefGoogle ScholarPubMed
18.Moritz, KM, Boon, WM & Wintour, EM (2005) Glucocorticoid programming of adult disease. Cell Tissue Res 322, 8188.CrossRefGoogle ScholarPubMed
19.Bard, JB (2002) Growth and death in the developing mammalian kidney: signals, receptors and conversations. Bioessays 24, 7282.CrossRefGoogle ScholarPubMed
20.Brenner, B & Rector, F (2007) The Kidney. Philadelphia, PA: W.B. Saunders Company.Google Scholar
21.Godfrey, K, Robinson, S, Barker, DJ et al. (1996) Maternal nutrition in early and late pregnancy in relation to placental and fetal growth. Br Med J 312, 410414.CrossRefGoogle ScholarPubMed
22.Ong, KK & Dunger, DB (2004) Birth weight, infant growth and insulin resistance. Eur J Endocrinol 151, Suppl. 3, U131U139.CrossRefGoogle ScholarPubMed
23.Ymer, SI & Herington, AC (1992) Developmental expression of the growth hormone receptor gene in rabbit tissues. Mol Cell Endocrinol 83, 3949.CrossRefGoogle ScholarPubMed
24.Holzenberger, M, Hamard, G, Zaoui, R et al. (2001) Experimental IGF-I receptor deficiency generates a sexually dimorphic pattern of organ-specific growth deficits in mice, affecting fat tissue in particular. Endocrinology 142, 44694478.CrossRefGoogle ScholarPubMed
25.Hirschberg, R & Kopple, JD (1989) Effects of growth hormone and IGF-I on renal function. Kidney Int Suppl 27, S20S26.Google ScholarPubMed
26.Liu, ZZ, Wada, J, Alvares, K et al. (1993) Distribution and relevance of insulin-like growth factor-I receptor in metanephric development. Kidney Int 44, 12421250.CrossRefGoogle ScholarPubMed
27.Bridgewater, DJ, Dionne, JM, Butt, MJ et al. (2008) The role of the type I insulin-like growth factor receptor (IGF-IR) in glomerular integrity. Growth Horm IGF Res 18, 2637.CrossRefGoogle ScholarPubMed
28.Schubring, C, Siebler, T, Kratzsch, J et al. (1999) Leptin serum concentrations in healthy neonates within the first week of life: relation to insulin and growth hormone levels, skinfold thickness, body mass index and weight. Clin Endocrinol (Oxf) 51, 199204.CrossRefGoogle ScholarPubMed
29.Schubring, C, Kiess, W, Englaro, P et al. (1997) Levels of leptin in maternal serum, amniotic fluid, and arterial and venous cord blood: relation to neonatal and placental weight. J Clin Endocrinol Metab 82, 14801483.CrossRefGoogle ScholarPubMed
30.Schwartz, MW, Baskin, DG, Bukowski, TR et al. (1996) Specificity of leptin action on elevated blood glucose levels and hypothalamic neuropeptide Y gene expression in ob/ob mice. Diabetes 45, 531535.CrossRefGoogle ScholarPubMed
31.Bell, AW & Ehrhardt, RA (2002) Regulation of placental nutrient transport and implications for fetal growth. Nutr Res Rev 15, 211230.CrossRefGoogle ScholarPubMed
32.Senaris, R, Garcia-Caballero, T, Casabiell, X et al. (1997) Synthesis of leptin in human placenta. Endocrinology 138, 45014504.CrossRefGoogle ScholarPubMed
33.Bispham, J, Gopalakrishnan, GS, Dandrea, J et al. (2003) Maternal endocrine adaptation throughout pregnancy to nutritional manipulation: consequences for maternal plasma leptin and cortisol and the programming of fetal adipose tissue development. Endocrinology 144, 35753585.CrossRefGoogle ScholarPubMed
34.Serradeil-Le Gal, C, Raufaste, D, Brossard, G et al. (1997) Characterization and localization of leptin receptors in the rat kidney. FEBS Lett 404, 185191.CrossRefGoogle ScholarPubMed
35.Beltowski, J, Jamroz-Wisniewska, A, Borkowska, E et al. (2004) Up-regulation of renal Na+, K+-ATPase: the possible novel mechanism of leptin-induced hypertension. Pol J Pharmacol 56, 213222.Google ScholarPubMed
36.Hall, JE (2003) The kidney, hypertension, and obesity. Hypertension 41, 625633.CrossRefGoogle ScholarPubMed
37.Rahmouni, K, Morgan, DA, Morgan, GM et al. (2005) Role of selective leptin resistance in diet-induced obesity hypertension. Diabetes 54, 20122018.CrossRefGoogle ScholarPubMed
38.Symonds, ME, Stephenson, T, Gardner, DS et al. (2007) Long-term effects of nutritional programming of the embryo and fetus: mechanisms and critical windows. Reprod Fertil Dev 19, 5363.CrossRefGoogle ScholarPubMed
39.Whorwood, CB, Firth, KM, Budge, H et al. (2001) Maternal undernutrition during early to midgestation programs tissue-specific alterations in the expression of the glucocorticoid receptor, 11beta-hydroxysteroid dehydrogenase isoforms, and type 1 angiotensin ii receptor in neonatal sheep. Endocrinology 142, 28542864.CrossRefGoogle ScholarPubMed
40.Williams, PJ, Kurlak, LO, Perkins, AC et al. (2007) Hypertension and impaired renal function accompany juvenile obesity: the effect of prenatal diet. Kidney Int 72, 279289.CrossRefGoogle ScholarPubMed
41.Bispham, J, Gardner, DS, Gnanalingham, MG et al. (2005) Maternal nutritional programming of fetal adipose tissue development: differential effects on messenger ribonucleic acid abundance for uncoupling proteins and peroxisome proliferator-activated and prolactin receptors. Endocrinology 146, 39433949.CrossRefGoogle ScholarPubMed
42.Gopalakrishnan, GS, Gardner, DS, Rhind, SM et al. (2004) Programming of adult cardiovascular function after early maternal undernutrition in sheep. Am J Physiol Regul Integr Comp Physiol 287, R12R20.CrossRefGoogle ScholarPubMed
43.Symonds, ME, Sebert, SP, Hyatt, MA et al. (2009) Nutritional programming of the metabolic syndrome. Nat Rev Endocrinol 5, 604610.CrossRefGoogle ScholarPubMed
44.Gopalakrishnan, GS, Gardner, DS, Dandrea, J et al. (2005) Influence of maternal pre-pregnancy body composition and diet during early-mid pregnancy on cardiovascular function and nephron number in juvenile sheep. Br J Nutr 94, 938947.CrossRefGoogle ScholarPubMed
45.Stein, CE, Fall, CH, Kumaran, K et al. (1996) Fetal growth and coronary heart disease in south India. Lancet 348, 12691273.CrossRefGoogle ScholarPubMed
46.Ogilvy-Stuart, AL, Hands, SJ, Adcock, CJ et al. (1998) Insulin, insulin-like growth factor I (IGF-I), IGF-binding protein-1, growth hormone, and feeding in the newborn. J Clin Endocrinol Metab 83, 35503557.CrossRefGoogle ScholarPubMed
47.Low, LC, Tam, SY, Kwan, EY et al. (2001) Onset of significant GH dependence of serum IGF-I and IGF-binding protein 3 concentrations in early life. Pediatr Res 50, 737742.CrossRefGoogle ScholarPubMed
48.Tannenbaum, GS, Guyda, HJ & Posner, BI (1983) Insulin-like growth factors: a role in growth hormone negative feedback and body weight regulation via brain. Science 220, 7779.CrossRefGoogle ScholarPubMed
49.Adelman, RD, Restaino, IG, Alon, US et al. (2001) Proteinuria and focal segmental glomerulosclerosis in severely obese adolescents. J Pediatr 138, 481485.CrossRefGoogle ScholarPubMed
50.Chagnac, A, Weinstein, T, Korzets, A et al. (2000) Glomerular hemodynamics in severe obesity. Am J Physiol Renal Physiol 278, F817F822.CrossRefGoogle ScholarPubMed
51.Praga, M, Hernandez, E, Herrero, JC et al. (2000) Influence of obesity on the appearance of proteinuria and renal insufficiency after unilateral nephrectomy. Kidney Int 58, 21112118.CrossRefGoogle ScholarPubMed
52.Rea, DJ, Heimbach, JK, Grande, JP et al. (2006) Glomerular volume and renal histology in obese and non-obese living kidney donors. Kidney Int 70, 16361641.CrossRefGoogle ScholarPubMed
53.Nakamura, M, Onoda, T, Itai, K et al. (2004) Association between serum C-reactive protein levels and microalbuminuria: a population-based cross-sectional study in northern Iwate, Japan. Intern Med 43, 919925.CrossRefGoogle ScholarPubMed
54.Ramkumar, N, Cheung, AK, Pappas, LM et al. (2004) Association of obesity with inflammation in chronic kidney disease: a cross-sectional study. J Ren Nutr 14, 201207.CrossRefGoogle ScholarPubMed
55.Hotamisligil, GS, Shargill, NS & Spiegelman, BM (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259, 8791.CrossRefGoogle ScholarPubMed
56.Cinti, S, Mitchell, G, Barbatelli, G et al. (2005) Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 46, 23472355.CrossRefGoogle ScholarPubMed
57.Fried, SK, Bunkin, DA & Greenberg, AS (1998) Omental and subcutaneous adipose tissues of obese subjects release interleukin-6: depot difference and regulation by glucocorticoid. J Clin Endocrinol Metab 83, 847850.Google ScholarPubMed
58.Massiera, F, Bloch-Faure, M, Ceiler, D et al. (2001) Adipose angiotensinogen is involved in adipose tissue growth and blood pressure regulation. FASEB J 15, 27272729.CrossRefGoogle ScholarPubMed
59.van Harmelen, V, Skurk, T, Rohrig, K et al. (2003) Effect of BMI and age on adipose tissue cellularity and differentiation capacity in women. Int J Obes Relat Metab Disord 27, 889895.CrossRefGoogle ScholarPubMed
60.Aubert, J, Darimont, C, Safonova, I et al. (1997) Regulation by glucocorticoids of angiotensinogen gene expression and secretion in adipose cells. Biochem J 328 (Pt 2), 701706.CrossRefGoogle ScholarPubMed
61.Hasdan, G, Benchetrit, S, Rashid, G et al. (2002) Endothelial dysfunction and hypertension in 5/6 nephrectomized rats are mediated by vascular superoxide. Kidney Int 61, 586590.CrossRefGoogle ScholarPubMed
62.Johnson, RJ, Alpers, CE, Yoshimura, A et al. (1992) Renal injury from angiotensin II-mediated hypertension. Hypertension 19, 464474.CrossRefGoogle ScholarPubMed
63.Abrass, CK, Raugi, GJ, Gabourel, LS et al. (1988) Insulin and insulin-like growth factor I binding to cultured rat glomerular mesangial cells. Endocrinology 123, 24322439.CrossRefGoogle ScholarPubMed
64.Berfield, AK, Andress, DL & Abrass, CK (2002) IGF-1-induced lipid accumulation impairs mesangial cell migration and contractile function. Kidney Int 62, 12291237.CrossRefGoogle ScholarPubMed
65.Saito, K, Ishizaka, N, Hara, M et al. (2005) Lipid accumulation and transforming growth factor-beta upregulation in the kidneys of rats administered angiotensin II. Hypertension 46, 11801185.CrossRefGoogle ScholarPubMed
66.Lauzier, MC, Page, EL, Michaud, MD et al. (2007) Differential regulation of hypoxia-inducible factor-1 through receptor tyrosine kinase transactivation in vascular smooth muscle cells. Endocrinology 148, 40234031.CrossRefGoogle ScholarPubMed
67.Lubbers, DW & Baumgartl, H (1997) Heterogeneities and profiles of oxygen pressure in brain and kidney as examples of the pO2 distribution in the living tissue. Kidney Int 51, 372380.CrossRefGoogle ScholarPubMed
68.Wiggins, JE, Goyal, M, Sanden, SK et al. (2005) Podocyte hypertrophy, “adaptation,” and “decompensation” associated with glomerular enlargement and glomerulosclerosis in the aging rat: prevention by calorie restriction. J Am Soc Nephrol 16, 29532966.CrossRefGoogle ScholarPubMed
69.Praga, M, Hernandez, E, Morales, E et al. (2001) Clinical features and long-term outcome of obesity-associated focal segmental glomerulosclerosis. Nephrol Dial Transplant 16, 17901798.CrossRefGoogle ScholarPubMed
70.Advani, A, Kelly, DJ, Advani, SL et al. (2007) Role of VEGF in maintaining renal structure and function under normotensive and hypertensive conditions. Proc Natl Acad Sci USA 104, 1444814453.CrossRefGoogle ScholarPubMed
71.Sebert, SP, Hyatt, MA, Chan, LL et al. (2009) Maternal nutrient restriction between early and midgestation and its impact upon appetite regulation after juvenile obesity. Endocrinology 150, 634641.CrossRefGoogle ScholarPubMed
72.Sharkey, D, Gardner, DS, Fainberg, HP et al. (2009) Maternal nutrient restriction during pregnancy differentially alters the unfolded protein response in adipose and renal tissue of obese juvenile offspring. FASEB J 5, 13141324.CrossRefGoogle Scholar
73.Chan, LL, Sebert, SP, Hyatt, MA et al. (2009) Effect of maternal nutrient restriction from early to midgestation on cardiac function and metabolism after adolescent-onset obesity. Am J Physiol Regul Integr Comp Physiol 296, R1455R1463.CrossRefGoogle ScholarPubMed
74.Zhou, YT, Grayburn, P, Karim, A et al. (2000) Lipotoxic heart disease in obese rats: implications for human obesity. Proc Natl Acad Sci USA 97, 17841789.CrossRefGoogle ScholarPubMed
75.Henegar, JR, Bigler, SA, Henegar, LK et al. (2001) Functional and structural changes in the kidney in the early stages of obesity. J Am Soc Nephrol 12, 12111217.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Diagrammatic representation of renal responses in offspring born to mid-gestation nutrient-restricted (NR) mothers compared to their control (C) group, as observed previously in sheep. Exposure to an obesogenic environment induced a series of hormonal responses triggered by changes in adipocyte responses to excess lipid accumulation leading to tissue remodelling and alterations in renal and related peripheral tissues. RAS, renin–angiotensin system; IGF, insulin-like growth factor.