Vous êtes sur la page 1sur 19

Ann. N.Y. Acad. Sci.

ISSN 0077-8923

A N N A L S O F T H E N E W Y O R K A C A D E M Y O F SC I E N C E S
Issue: The Year in Diabetes and Obesity

Early life nutrition and metabolic programming


Denise S. Fernandez-Twinn and Susan E. Ozanne
Department of Clinical Biochemistry, University of Cambridge, Metabolic Research Laboratories, Institute of Metabolic
Sciences, Addenbrookes Hospital, Cambridge, United Kingdom

Address for correspondence: Denise S. Fernandez-Twinn, Ph.D., Department of Clinical Biochemistry, University of
Cambridge, Metabolic Research Laboratories, Institute of Metabolic Sciences, Addenbrookes Hospital, Cambridge, United
Kingdom. df220@cam.ac.uk

Research investigating the early programming of adult metabolic disease has in recent years provided much mecha-
nistic insight into how the early environment impacts on long-term health. It includes studies addressing the roles
of intrauterine nutrient availability, which is determined by maternal nutrition, maternal exposure to oxygen, toxic
events, and infection; the placental interface; and also the early postnatal environment. This review will explore
the epidemiological evidence for programming of metabolic disease and provide an overview of the various studies
using animals to model metabolic phenotypic outcome. It will also discuss evidence for the proposed molecular
mechanisms and the potential for intervention.

Keywords: metabolic syndrome; insulin resistance; early life nutritional programming; insulin signaling

The metabolic syndrome phenotype as fuel. Type 2 diabetes is an age-related disease,


generally occurring in individuals over the age of
Metabolic syndrome presents as a combination of a
40; however, in some ethnic groups, such as South
variety of symptoms, which include fasting hyper-
Asian and African-Caribbean, it often appears af-
glycemia, impaired fasting glucose, impaired glu-
ter the age of 25. An increasing number of children
cose tolerance or insulin resistance, systemic inflam-
are also being diagnosed with the condition. More
mation, hypertension, central obesity (also known
than 2.6 million people have been diagnosed with
as visceral, male-pattern or apple-shaped adiposity),
the disease, and the number is predicted to rise to 4
decreased HDL cholesterol, and elevated triglyc-
million by 2025.2
erides. While there is some disagreement regarding
causality and the relative importance of each of these The concept of programming
criteria in the formal definition of the metabolic syn-
drome, it is undisputed that the extent of this disease From the moment of conception, an organisms fu-
is far-reaching, with prevalence in the US estimated ture physiological and even psychological attributes
at 25% of the population.1 Equally important is the are being defined by the conditions in which it is
associated risk for cardiovascular disease. growing. Early blastocyst cell division, implantation
Of the many features outlined, impaired glu- into the maternal endometrium, trophoblastic inva-
cose homeostasis is one of the major components sion, and subsequent patterning are all orchestrated
of metabolic syndrome. Islet beta cell dysfunction by the quantity and quality of nutrition obtained
and/or resistance to the actions of insulin in skeletal through the maternal-fetal interface, that is, the pla-
muscle, liver, and adipose tissue are instrumental centa. Fetal demands that are not met by maternal
in the development of type 2 diabetes. Insufficient nutrient supply can hinder the fetal growth trajec-
insulin production by the beta cells of the islets and tory,3 which could have consequences for long-term
its secretion results in beta cell dysfunction while health. Conversely, maternal substrate availability
insulin resistance is characterized by incomplete up- that exceeds fetal requirements can also superim-
take and storage of glucose and amino acids for use pose deviations in normal growth patterns and thus

doi: 10.1111/j.1749-6632.2010.05798.x
78 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 
c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

confer disease outcomes over the life course. Fetal est and highest birth weight individuals.1517 An
programming describes the in-utero phenomenon increased risk of reproductive disorders such as
through which these deviations are hard-wired into polycystic ovary syndrome at adolescence, which
every cell in the organism, and thereby determines is related to premature adrenarche in girls18 as
future disease susceptibility. Catastrophic events in well as hypospadia and cryptorchidism in boys,19
history such as the Dutch Hunger Winter and the is also linked with low birth weight. A few stud-
Leningrad Siege, during which rations were strictly ies have suggested that the increased risk of end-
imposed on all sections of the population including stage renal disease20 in low birth weight individuals
pregnant and nursing mothers, presented the op- stems from impaired nephron development result-
portunity to study associations with chronic adult ing in reduced glomerular number and increased
disease in babies born around the period. These glomerular volume.21 Finally, while links between
studies stress the importance of the pre- and early lower neuropsychological performance22 and inci-
postnatal environment in growth and development, dence of schizophrenia23 and low birth weight have
and that the timing of an insult or deviation from been shown, some of these observations have been
the norm is as important as the insult itself in deter- suggested to be confounded by maternal psychi-
mining (a) the organ system(s) affected and (b) the atric disorders and paternal age,24 with maternal
onset and severity of disease outcome. schizophrenia being associated with an increased
Since these initial studies, a large number of risk of low birth weight. Schizophrenia is also related
epidemiological studies have consistently demon- to increased risk of febrile seizures, which may be ge-
strated the relationship between poor fetal and netically transmitted through either parent.25 Fur-
early postnatal growth and the development of thermore, some factors relating to low birth weight
adult diseases. Using proxy indices based on records such as prenatal exposure to maternal smoking, ma-
of birth weight, length, abdominal and head cir- ternal alcohol and coffee consumption, maternal
cumference, placental weight, and various relative binge drinking, maternal stress, and preeclampsia
indices as markers of in-utero growth, numerous were shown to play little or no causal role in the
epidemiological studies have explored the relation- etiology of febrile seizures.26
ships between early human growth and suscepti-
bility to adult disease. Since there is still no single Early growth and metabolic disease:
comprehensive test to identify if a newborn has at- current hypotheses
tained its full growth potential in-utero, these crude The relationship particularly between poor early
measures are still widely used. Hence, strong as- growth and the development of type 2 diabetes and
sociations between low birth weight and increased the metabolic syndrome is robust, as it has been ob-
risk of metabolic disorders are well documented. served in a wide range of populations and in many
Cardiovascular disease,4 type 2 diabetes,5 central different ethnicities.27,28 The mechanisms underly-
adiposity,6 abnormal lipid metabolism and hy- ing this relationship and the relative contributions
pertension,7 and increased risk of death from is- of genes and the environment and the interactions
chemic heart disease8,9 have all been consistently between the two, however, are much debated.
linked to birth weight. The associations with acute
myeloid leukemia10 and certain cancers such as Genes: fetal insulin hypothesis
testicular cancer11 are much weaker (probably at Rare mutations in the glucokinase gene have been
least in part due to low sample number and effect shown to be associated with low birth weight and the
size), while associations with esophageal adenocar- development of a rare monogenic form of diabetes,
cinoma12 is thought to be explained by increased maturity onset diabetes of the young (MODY).29
gastroesophageal reflux during infancy among in- This data formed the basis of the Fetal Insulin
fants born preterm and/or small for gestational age Hypothesis that suggests the relationship between
(SGA). Birth weight associations with breast cancer birth weight and type 2 diabetes arises because of
are seemingly contradictory, as most studies show polymorphisms/mutations in genes such as those
increased risk with increasing birth weight,13,14 implicated in insulin secretion causing both re-
while some other studies regarded risk as increased duced fetal growth and increased risk of type two
in a J-shaped association, that is, in both the low- diabetes. Since this finding, rapid advances in

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 79
Early life programming Fernandez-Twinn & Ozanne

sequencing technology have facilitated a revolution meets with nutritional abundance in later life. Fur-
in genome wide association studies and large Eu- ther studies supporting this hypothesis followed. In
ropean cohort studies have identified genetic vari- the Dutch Hunger Winter study, reduced glucose
ants associated with birth weight.30 More recently, tolerance was observed in adults who were in-utero
one study demonstrated the association of Single at the time of strict food rationing towards the end
Nucleotide Polymorphisms within adenylate cy- of World War II.33 This effect was most marked in
clase type 5 (ADCY5), prospero homeobox pro- individuals who were in their third trimester during
tein 1 (PROX1), glucokinase (GCK ), glucokinase the period of the famine, which added a temporal di-
regulator (GCKR) and diacylglycerol kinase beta- mension to the severity of disease. Further strong ev-
transmembrane protein 195 (DGKB-TMEM195) idence for the importance of the fetal environment
with Type-2 diabetes31 and in the second, vari- came from studies of twins,3436 which showed dis-
ants at the ADCY5, vacuolar protein sorting 13 ho- cordance for type 2 diabetes, with the diabetic twin
molog (VPS13C), GCKR and transcription factor having a significantly lower birth weight than the
7-like (TCF7L2) loci correlated with 2-h glucose. nondiabetic co-twin regardless of zygosity.36 Many
These provide evidence that the well-described as- more reviews have since supported the association
sociation between low birth weight and subsequent between low birth weight and adult glucose and in-
type 2 diabetes has a genetic component, distinct sulin metabolism.37,38 In recent years, Gluckman &
from the proposed role of programming by maternal Hanson proposed a related hypothesis, that is, the
nutrition. predictive adaptive response hypothesis (PAR),39
which states that the fetus makes adaptive metabolic
Maternal environment and the developmental adjustments based on its in-utero environment that
origins of health and disease predicts the environment into which it is likely to
Nutrient restriction. The in-utero environment is be born in order to gain a competitive advantage
extremely labile to maternal influence, which could after birth. The effects of the maternal environment
therefore play a crucial role in fetal growth. The ev- on the developmental path of the offspring are thus
idence for this comes from epidemiological studies to gain evolutionary advantage by epigenetic means
demonstrating associations between various differ- and transmission via soft inheritance40 to future
ent types of maternal exposures on birth weight, generations.
with adverse consequences for metabolic disease.
Hypoxia. Fetal hypoxia correlates with cardiovas-
The earliest epidemiological study linking poor fetal
cular disease in later adult life; evidence for this
growth and subsequent development of type 2 dia-
comes from studies on the highland inhabitants
betes was the observation by Hales and colleagues,5
of Bolivia, when Giussani and colleagues showed
which found that among men in their sixties, those
that a low oxygen tension was associated41 with low
who had lower birth weights and weights at 1 year
birth weight and altered body shape at birth. Epi-
were more likely to develop poor glucose tolerance
demiological studies in other populations living at
and type 2 diabetes. These findings led to the pro-
high altitudes in Saudi Arabia42 confirmed the ef-
posal of the thrifty phenotype hypothesis,32 which
fects on low birth weight. Adaptations in multigen-
suggests that fetal malnutrition may induce physio-
erational high altitude populations (e.g., Andeans
logical and/or metabolic adaptations to ensure nu-
and Tibetans), however, seem to permit higher uter-
trient supply to the most vital organs (such as the
ine artery blood flows and protect against altitude-
brain) at the expense of other organs (e.g., the en-
associated IUGR.43
docrine pancreas). Furthermore, such adaptations
during critical windows of development may per- Maternal anaemia/micronutrient deficiency. The
manently reset (e.g., reduced beta cell mass) or prevalence of anaemia in pregnant women is es-
program fetal metabolism in order to enhance its timated at 41.8% globally,44 and is particularly a
chances of survival in what it expects to be a poor nu- problem in the developing world. Both maternal
tritional environment (Fig. 1). This program of phe- anaemia45 and iron deficiency are associated with
notypic adjustment selectively maximises metabolic infant and childhood mortality. Epidemiological
thrift; however, this advantage becomes detrimen- observations demonstrate a relationship between
tal to the programmed individual/offspring when it maternal anemia and preterm delivery and low

80 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

Figure 1. A schematic representation of the structural and molecular programming effects of a suboptimal in-utero environment
on early growth and subsequent development of the metabolic syndrome.

birth weight46 with reduced placental vasculariza- colleagues reported a small increase in birth weight
tion and deregulation of fetal insulin-like growth and a reduction in the prevalence of low birth weight
factor 1 (IGF-1) and corticotropin-releasing hor- (LBW) of about 10% in pregnancies supplemented
mone (CRH). It is suggested that effects of iron with iron-folic acid.50
deficiency or hypoxia on the fetus may be me-
diated by sympathetic activation through raised Maternal stress. In humans, glucocorticoids are
norepinephrine, and this may serve as a stimulus administered during late gestation to treat neona-
for cortisol and CRH release.47 The fetus is nor- tal respiratory morbidity and maternal asthma. The
mally protected against excessive cortisol exposure side effects of this treatment are growth retardation
through placental 11--hydroxysteriod dehydroge- and reduced birth weight.51 It is hypothesized that
nase (11HSD2), which converts cortisol to inactive prenatal glucocorticoids or stress mediates the pro-
cortisone, however, evidence from animal studies gramming of the hypothalamopituitaryadrenal
suggest that copper, zinc or vitamin E deficiency re- (HPA) axis as an excess of glucocorticoids may
duce the expression of this enzyme.48 At present, be associated with hypertension and glucose intol-
associations between maternal iron deficiency and erance.52 Early studies by Vallee and colleagues53
metabolic consequences for offspring are indirect, showed that offspring of prentally stressed rat dams
coming from studies of maternal supplementation. had increased basal blood glucose levels, ate less,
Christian and colleagues recently reported their and weighed less than their control counterparts.
findings on a randomized trial demonstrating the Later studies54 reproduced this finding that is, ma-
effects of maternal antenatal/postnatal supplemen- ternal stress reduced body, adrenal, and pancreas
tation with iron-folic acid plus Vitamin A at allevi- weight as well as plasma corticosterone and glu-
ating mortality,49 while a meta-anaylsis by Fall and cose levels in term fetuses, whilst aged male rats

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 81
Early life programming Fernandez-Twinn & Ozanne

(24 months of age) demonstrated hyperglycaemia, ther studies across many continents have confirmed
glucose intolerance, and decreased basal leptin lev- the importance of postnatal growth in the program-
els. Furthermore, food intake following a fast was ming of metabolic syndrome in South African,60
increased. Thus, an adverse glucocorticoid environ- Finnish,61 and Indian62 cohorts, with the findings
ment in-utero induced a long-lasting disturbance in that low birth weight babies that underwent rapid
feeding behavior and metabolic dysfunction. postnatal weight gain demonstrated the worst glu-
The underlying mechanism directing glucocor- cose tolerance and were therefore the most likely to
ticoid excess is thought to be physiological varia- develop type 2 diabetes in adulthood. The worsen-
tions in placental 11HSD2 activity. Altered pla- ing of glucose tolerance in children who were born
cental glucocorticoid metabolism is associated with small but then had the highest weight gain corre-
nutrient restriction and hypoxia, which lead to both lated to the greatest beta cell secretory response,
intrauterine growth restriction (IUGR) and fetal and those who were the most insulin resistant.63 In
programming. Placental 11HSD2 expression and contrast, children born lighter and whose weight
activity is regulated in a cell- and gestational age remained low had the lowest beta cell secretory re-
specific manner in both humans and baboons.55 sponse. Both the immediate postnatal period and
In humans, mutations in the 11HSD2 gene have the first few years of childhood are attributed as the
been associated with low birth weight. In rodents, phases during which accelerated growth has added
reduced 11HSD2 activity, and increased fetal cor- detrimental effects for metabolic disease.64
tisol levels are also associated with IUGR.56 In sheep,
Gestational diabetes. Gestational diabetes (GDM)
maternal nutrient restriction for more than 45 days
can occur due to a pre-existing diabetic state or the
significantly decreased placental 11HSD2 in ad-
development of glucose intolerance during preg-
dition to causing IUGR.57 This reduced 11HSD2
nancy. Depending on the severity of GDM, babies
expression and activity in preeclampsia58 may be
may either be macrosomic or growth restricted and
due to a sustained placental hypoxia due to defi-
it is thought that the placenta plays an important
cient trophoblast invasion in the first trimester.
role since it has been shown that placental size in
Mutations in the 11HSD2 gene cause low birth
women diagnosed with GDM is inversely related to
weight, and these infants go on to have higher
protein intake.65 Epidemiological studies on cases of
plasma cortisol levels throughout their adult life.
diabetes in the Netherlands,66 Malta,67 Pima Indi-
Phillips and colleagues also observed this association
ans,68 and African-Americans69 have demonstrated
in three different populations in Adelaide, Australia,
a U-shaped relationship with birth weight, so that
and in Preston & Hertfordshire in the UK. They
macrosomia poses as much a risk for metabolic dis-
found that higher cortisol concentrations within
ease as low birth weight. There is also a higher sus-
the normal range were associated with raised blood
ceptibility for diabetes in descendants of diabetic
pressure, impaired glucose tolerance, insulin resis-
great grandmothers via the maternal line, with a
tance, and raised serum triglyceride levels, suggest-
higher prevalence of impaired glucose tolerance,
ing intrauterine programming of the HPA axis as a
type 2 diabetes and gestational diabetes occurring
mechanism underlying the association between low
in children whose mothers had diabetes during that
birth weight and the insulin resistance syndrome in
pregnancy.68 Gestational diabetes per se is an induc-
adult life.59
ing factor for impaired glucose tolerance and ges-
tational diabetes in the next generation; the altered
Mismatch between poor in-utero growth and post-
metabolic milieu of the diabetic pregnancy causing
natal growth. The influences of restricted fetal
permanent defects in glucose homeostasis in the off-
growth need to be examined with reference to the
spring leading to the development of diabetes later
equal importance of postnatal nutrition, another
in life. This forms the basis of a mechanism by which
component of the Thrifty Phenotype hypothesis.
gestational diabetes passes from one generation to
When cases in the epidemiological studies of both
the next.
the Hertfordshire and Dutch Hunger Winter were
examined further, it was found that the individuals Maternal obesity. Pregnancies during which
with the worst glucose tolerances were those who mothers are obese or overeat70 can also result
were born small and became obese as adults5,33 Fur- in increased infant birth weight and increased

82 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

susceptibility to metabolic and related diseases in ing nutrient and gas transport, and by the synthesis
adult life. Maternal obesity and gestational weight and secretion of steroid and peptide hormones. How
gain significantly impact on maternal metabolism well it does this is dependent on vascular develop-
with effects on maternal insulin sensitivity, glu- ment and blood flow and by growth and differenti-
cose tolerance and lipid metabolism, and the de- ation of the trophoblast, which contains receptors,
livery of either a SGA or a large for gestational age transporters, and enzymes.84,85 Epidemiological ev-
baby.70 Pregravid BMI > 30 was shown to be a very idence has linked low birth weight and low pla-
strong predictor of childhood obesity body mass in- cental weight to fetal programming. Alterations in
dex (BMI).71 In addition, pregnancy weight gain72 expression of glucose and amino acid transporters
and consumption of six glasses of cows milk per in the placenta have been reported in pregnancies
day during mid-pregnancy73 were also shown to be complicated by diabetes, IUGR, and preeclampsia86
independent determinants of macrosomia. In the with hyperglycaemia in the first trimester result-
latter study, increased birth weight was related to ing in up-regulation of glucose transporters, lead-
the increased protein intake in the milk rather than ing to accelerated fetal growth in late gestation. Nu-
fat. BMI and pregnancy weight gain prior to preg- merous excellent studies by Fowden and colleagues
nancy are also strong predictors of gestational dia- have meticulously detailed the morphological and
betes, which could be a mechanism linking maternal functional adaptations of the placenta in adverse in-
obesity and over-nutrition to high birth weight and trauterine conditions.87 Depending on the model of
metabolic disease consequences for offspring.74 IUGR and the severity of the insult, the consensus
Healthy pregnancy is associated with lipid mo- from the literature is that placental weight is gener-
bilization (increases in plasma cholesterol and ally reduced in parallel with fetal weight; however,
triacylglycerols), changes in insulin sensitivity (a placental efficiency, as assessed by grams fetus per
gestational increase in insulin resistance) and im- gram placenta, is largely variable. This suggests that
provement in endothelial function.75 In mater- placental efficiency is determined in part by genet-
nal obesity, there is an exaggerated lipid response, ics, and in part, by its response to the pregnancy
greater degree of insulin resistance76 and less environment. The response to maternal under nu-
improvement in endothelium-dependent microvas- trition for example, is an increase in placental ef-
cular function.77 The relative endothelial dysfunc- ficiency during the early growth spurt (gestational
tion in obese pregnancy is in part explained by al- days 1516), and then a decline in efficiency closer
tered levels of plasma cytokines (interleukin-6 and to term.88,89 However, the severity of the insult also
-10) with obesity.78 Lipid peroxides are increased affects placental efficiency as seen in cases of more
in healthy pregnancy,79 but few findings are avail- severe restriction that is, a 4% protein diet during
able on oxidative stress in obese pregnancy. An as- pregnancy, where placental efficiency was reduced
sociation between maternal BMI and also mater- at day 21 gestation86 compared to intake of a 8%
nal cholesterol levels and the oxidizability of low protein diet, which resulted in increased placental
density lipoprotein (LDL) has been observed.80 In efficiency.90,91 Morphologial adaptations to surface
nonpregnant type 2 diabetics, high plasma oxys- area, thickness of the barrier between the maternal
terol levels are particularly observed when plasma and fetal circulations, and the density and archi-
LDL levels are above approximately 3.6 mmol/l, a tecture of the vasculature contribute to improved
level exceeded in overweight and obese women.81 efficiency and these are species dependent. How-
Although the literature on oxysterols during preg- ever, in addition to this are transporter-mediated
nancy is extremely limited, gestational increases and processes which are determined by abundance, ac-
further increases with maternal diabetes have been tivity and localization of transporters as well as sim-
reported.82 ple and facilitated diffusion which is affected by the
materno-fetal concentration gradient across the pla-
Role of the placenta
centa.92 The expression of a range of glucose (Slc2a1
Placental dysfunction is implicated as a major cause and 3, GLUT1, GLUT3) and amino-acid trans-
of human IUGR and programming of adult car- porters (Slc38a2 and 4) have been found to be up-
diovascular and metabolic disease.83 The placenta regulated in various models of IUGR (as reviewed by
regulates fetal growth and development by facilitat- Fowden et al.87 ). What drives these morphological

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 83
Early life programming Fernandez-Twinn & Ozanne

and functional modifications in the placenta is im- schematic in Figure 1. Large animal models of IUGR
portant as these could potentially act as biomarkers have also been studied in depth,103 and although
of IUGR, and may also serve as a route to interven- these will not be discussed here, it is noteworthy
tion. Concentrations of maternal glucocorticoids, that they share many similarities with the rodent
IGF-1, leptin, and insulin have all been found to models that will be deliberated in greater detail.
be associated with IUGR.91,93,94 In the mouse, dele-
Rodent models and underlying mechanisms
tion of the Igf2 gene leads to placental and fetal
Maternal calorie restriction. Total maternal calo-
growth retardation, and conversely, overexpression
rie restriction to 50% of ad libitum intake during the
causes overgrowth.95 All these are likely candidates
last week of pregnancy results in impairment of beta
for modifying placental efficiency.
cell development.104 Continued calorie restriction
Martin and Sutherland96 showed that embryos
during lactation results in a permanent reduction
cultured in medium lacking amino acids were un-
in beta-cell mass and number, and impaired glucose
able to form trophoblast cell outgrowths necessary
tolerance in the offspring.105 A more severe food re-
for implantation and that mTOR is required for the
striction to 30% of ad libitum intake resulted in
initiation of this trophectoderm protrusive activity.
systolic hypertension and increased fasting insulin
Wen and colleagues later showed that mTOR func-
concentrations, hyperphagic behavior, and obesity
tions as a placental growth signaling sensor.97 Hu-
in the offspring.106 When treated with IGF-I, this
man studies are in agreement as mammalian target
phenotype was ameliorated107 while neonatal lep-
of rapamycin (mTOR) protein is expressed in the
tin administration from day 3 to day 13 of postnatal
transporting epithelium of the human placenta. In
life normalized caloric intake, locomotor activity,
these studies,98 the protein expression of placen-
body weight, fat mass, and fasting plasma glucose,
tal phospho-S6K1 (Thr-389), a measure of active
insulin, and leptin concentrations in adult life.108
mTOR signaling was markedly reduced in IUGR
Postnatal consequences for the HPA axis include re-
placentas.
duced adrenal weight, reduced basal corticosterone
One other means of regulating placental func-
as well as reduced glucocorticoid receptor (GR) and
tion is through oxidative stress. As the placenta is a
CRH mRNA levels. Similar findings were observed
mitochondria-rich tissue, it generates reactive oxy-
by Dumortier and colleagues109 with fetuses of dams
gen species (ROS), even in a normal pregnancy,
fed a low energy diet demonstrating 3356% reduc-
which consigns it to a state of continuous oxida-
tion in beta cell mass and increased corticosterone
tive stress.99 This is increased in pregnancies com-
levels. Furthermore, the number of cells producing
plicated by preeclampsia, IUGR, and pregestational
neurogenin 3 (NEUROG3) or pancreatic and duo-
diabetes as measured by increased markers of pro-
denal homeobox gene 1 (PDX-1) was also reduced.
duction of ROS and decreases in antioxidant de-
fences.100 The defective trophoblast invasion seen Maternal protein restriction. The low protein (LP)
in preeclampsia or IUGR is thought to lead to a model of programming is the best characterized
relative hypoxia,101 which then leads to a height- of all developmental programming models. The
ened inflammatory response and thereby increases Hoet group first implemented the model,110 which
oxidative and nitrative stress. This is hypothesized corresponds with the levels of protein deprivation
to alter placental function via covalent modification observed in countries with poor socio-economic
of protein structure and function102 and hence fetal conditions. The LP diet, which is fed only to the
growth and development. rat mothers, consists of 8% casein protein and is
isocaloric with the control diet, which contains 20%
Animal models of programming
casein protein by comparison. Offspring suckle on
The following section provides a snapshot of re- their respective mothers milk and then are weaned
cent and current lines of research that have aimed onto standard chow, which contains 20% protein.
to dissect the physiological and molecular mech- The offspring are growth restricted, as characterized
anisms underlying the programming of metabolic by low birth weight and reduced placental weights,
disease. The major rodent animal models used to immediately demonstrating its relevance for study-
identify mechanisms of metabolic programming ing human growth restriction. Others have since
and observed offspring phenotypes are outlined in a replicated this gross morphological phenotype.91,111

84 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

At the tissue level, neonate beta cell proliferation received 20% protein in-utero and were suckled by
and islet size was reduced,112 and islet vasculariza- LP mothers, lived longer and contrary to the recu-
tion was also reduced.109 Our group and others have perated and control mice, were protected from the
extensively studied the adult phenotype also. LP off- detrimental effects of the obesogenic diet to reduce
spring at first show an improvement in their glucose longevity.123
tolerance up to 3 months of age. They undergo an The metabolic effects of catch-up growth have
age-dependent loss of glucose intolerance such that also been studied in different models. In mice off-
by 15 months of age, they have impaired glucose spring that were exposed to 50% food restriction
tolerance and insulin resistance and finally, frank in-utero followed by catch-up growth, increased
diabetes by 17 months of age.113 The insulin resis- adiposity was primarily associated with lipogene-
tance is associated with a reduced expression of key sis rather than adipogenesis.124 However, in a study
insulin signaling proteins, including the p110 cat- of forced catch-up growth following fetal protein re-
alytic subunit of phosphatidyl inositol 3-kinase in striction that showed similar increases in relative fat
adipose tissue114 and protein kinase C- in skele- mass and adipocyte hypertrophy, along with hyper-
tal muscle.115 The further relevance of this model glycemia, hypercholesterolemia, and hyperleptine-
to humans at the molecular level is the strikingly mia, Bol and colleagues125 reported a contradictory
similar profile of insulin signaling protein expres- down-regulation in mRNA expression of enzymes
sion in muscle and adipose tissue from LP offspring involved in lipogenesis, although there was a con-
compared to those in tissues from humans with a comitant up-regulation of proteins involved in fatty
low birth weight.116,117 That the differences in pro- acid uptake that could contribute to hypertrophy of
tein expression occur prior to development of in- fat cells.
sulin resistance, suggests that they are not conse- In a similar model, Sutton and colleagues126
quences of hyperinsulinemia or hyperglycemia but found abnormal daily rhythms in food intake and
play a role in determining future susceptibility to in- metabolism, increased lipogenesis, and inflamma-
sulin resistance, cardiovascular disease, and type 2 tion prior to increased obesity. At the molecular
diabetes. level, arrhythmic expression of circadian oscillator
genes in brain, liver, and muscle was evident as early
Catch-up growth. The respective roles of the preg- as 8 weeks of age (preobese); in particular the expres-
nancy and lactation period have been dissected by sion of the clock-associated nuclear receptor and
imposing the LP diet only during either of these pe- transcription repressor Rev-erb was reduced, with
riods through crossing-over techniques in rodents. concordant increased expression of its downstream
The offspring of rat dams fed a LP diet (8%) dur- targets Bmal1 and Per2 in liver and muscle. This
ing pregnancy have a 15% reduction in birth weight disruption in circadian rhythm is a potential mech-
compared to control offspring of dams fed a 20% anism for altered feeding behavior and substrate
diet. If these offspring are then cross-fostered to 20% metabolism, which may contribute to an increased
protein fed dams from birth, they undergo rapid diet-induced obesity and insulin resistance by 20
catch-up growth during the lactation period. The weeks of age. It remains unclear, however, due to the
most startling consequence was a 25% reduction in lack of a group that was not cross-fostered at birth,
life span in the recuperated male rats.118,119 The whether this disruption is programmed in-utero or
major cause of death in male rats is reported to during catch-up growth.
be kidney failure and consistent with this, reduced
longevity in the recuperated animals was associated Maternal micro-nutrient restriction. Early studies
with shorter kidney telomeres,119 although shorter in rats showed that insulin and glucagon levels in the
islet telomeres and increased markers of cell senes- fetus were affected by maternal zinc deficiency.127
cence have also been observed in these animals.120 More recent studies have shown offspring of zinc
Similar findings were observed in mice121,122 and restricted dams weighed less than control offspring
these studies also revealed an interaction with the at birth and weaning and up till 6 months of age.128
postweaning diet such that an obesity-inducing diet Their percentage of body fat was increased with a
further reduced the longevity of both control and concomitant decrease in lean mass. Fasting plasma
recuperated mice. Conversely, mice offspring that insulin levels in both male and female offspring at

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 85
Early life programming Fernandez-Twinn & Ozanne

6 months of age were lower compared to controls, Maternal glucocorticoid exposure. Prenatal glu-
which was related to a decreased glucose-induced cocorticoids or stress are hypothesized as a mech-
insulin secretion in females. anism linking reduced fetal growth with adult
Maternal iron restriction in a rat model has been metabolic syndrome disease. This has been tested in
shown to reduce birth weight and to elevate off- rats where the treatment of pregnant rats with dex-
spring blood pressure. This was suggested to be amethasone resulted in reduced birth weight, with
due to a deficit in nephron number.129 As with subsequent catch up in weight compared to controls
the protein restriction model, glucose tolerance was by weaning.139 In adulthood, offspring were hyper-
improved at 3 months of age, however, an age- tensive and hyperglycemic, concurrent with an in-
related loss of glucose tolerance was observed in creased HPA axis activity and behavior reminiscent
14-months-old rats, and it was suggested that this of anxiety.140 The underlying mechanism directing
may deteriorate with advancing age.130 glucocorticoid excess is thought to be physiological
variations in placental 11HSD2 activity.141 In hu-
Maternal hypoxia. Illsley and colleagues have re- mans, 11HSD2 gene mutations result in low birth
cently hypothesized that hypoxic conditions induce weight with consequences for higher plasma cortisol
metabolic reprogramming in the placenta such that levels in adult life, a clear programming effect on the
mitochondrial oxygen consumption is reduced and HPA axis. The effects are apparently gender-specific
anaerobic glucose consumption increases, thus al- such that males exhibit a metabolic phenotype139
tering the mix of fuel substrates available for trans- whereas females are hypertensive and have an acti-
fer to the fetus.131 Placental oxygen has been shown vated reninangiotensin system.142
to be preserved under maternal hypoxia;132 how-
Gestational diabetes. Experimental gestational di-
ever, this may be at the cost of less glucose availabil-
abetes may be induced by streptozotocin treatment
ity, leading to hypoglycemia-mediated fetal growth
of pregnant dams. This chemical destruction of
restriction.
islet beta cells of the pancreatic islets can be reg-
ulated such that a low dose induces mild gesta-
Intauterine artery ligation. It is now well estab-
tional diabetes resulting in fetal hyperinsulinemia
lished that a large proportion of human intrauter-
and macrosomia. Conversely, a high dose causes
ine growth retardation is due to impaired nutrient
severe maternal hyperglycemia resulting in hyper-
perfusion through the placenta.133 Both unilateral
stimulation of fetal islets followed by beta cell de-
and bilateral uterine artery ligation models of pla-
granulation, disorganization, and finally secretory
cental insufficiency replicate the phenotype of re-
failure. The result is a low birth weight pup.143 One
duced offspring birthweight and observations have
significant finding was that both extremes of birth
included an altered development of the pancreas,
weight converged on insulin secretory function and
with offspring at birth having a reduced beta cell
impaired glucose metabolism. Offspring of both
mass that persists into adulthood.134 This leads to
the mild and severely diabetic streptozotocin mod-
the development of type 2 diabetes at 26 weeks
els subsequently develop gestational diabetes them-
of age.135 Other effects of this model include re-
selves,144 which is further transmissible to the third
duced nephron number and impaired renal func-
generation.
tion136 and a 20% decrease in glomerular number
with compensatory glomerular enlargement associ- Maternal high fat feeding/over-nutrition. With
ating with an increased proteinuria.137 The effects of the prevalence of obesity rising to over a third
uterine ligation are transmitted to the second gen- of the population in some developed nations, it
eration as seen in female offspring of uterine ligated has become increasingly relevant and important
dams, which have reduced beta cell mass and be- to study models of obesity and particularly the
come diabetic during pregnancy, whereupon their consequences of obesity on the health of future gen-
offspring, the second generation, are heavier at birth erations. Recent years have seen much study dedi-
and remain heavy throughout life, develop insulin cated to the characterization of the phenotype of ro-
resistance very early in life, demonstrate defects in dent models of maternal obesity. One of the first of
insulin secretion from as early as 5 weeks of age and these demonstrated the development of hyperpha-
by 26 weeks of age are overtly diabetic.138 gia, adiposity, hypertension, and insulin resistance

86 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

in the offspring between 3 months and 6 months ate nucleus explants from d4-old neonatal mice.151
of age.145 Further work showed an attenuation In-vivo, daily leptin administration to d4 leptin defi-
of leptin-induced appetite suppression and phos- cient ob/ob mice was able to restore arcuate nucleus
phorylation of signal transducer of transcription 3 projections to the paraventricular nucleus to wild-
(STAT3) in the arcuate nucleus (ARC) and a re- type levels.
duction in AgRP-immunoreactivity in the hypotha-
lamic paraventricular nucleus (PVH) at postna- Molecular mechanisms
tal Day 30. These differences are therefore present Oxidative stress and mitochondrial mechanisms.
prior to the hyperphagic response and suggests lep- It is now widely known that oxidative damage
tin resistance as a potential mechanism.146 Tissue accelerates telomere shortening and thus cellular
specific insulin resistance and non-alcoholic fatty aging or senescence. Mitochondria, as the principal
liver disease (NAFLD) in liver has been observed energy generators (ATP) of the cell, generate large
in offspring challenged with a high fat diet post- amounts of ROS, such as superoxide anions and
weaning.147 In skeletal muscle, the expression of hydroxyl radicals and are therefore the primary me-
molecules involved in insulin signaling were altered diators of cellular oxidative damage. These oxygen
and mitochondrial-linked complex II-III activity radicals are capable of damage to protein, lipids,
was decreased, which could contribute to a state and DNA. Disruption in mitochondrial electron
of insulin resistance.148 transport chain (ETC) enzymes can lead to further
ROS generation,152 which in turn can contribute to
Other mechanisms
accelerated telomere length shortening and cellular
Permanent structural changes senescence under physiological conditions.153 Islets
Environmental factors at critical periods of devel- isolated from aged rat offspring (15 months) of
opment could have long-term phenotypic conse- dams protein restricted during pregnancy and
quences through permanent structural changes in lactation (LP model),154 demonstrate increased
key organs. Thus, inappropriate concentrations or xanthine oxidase expression, which is indicative of
a temporal shift in a particular nutrient or hor- increased oxidative-stress. Furthermore, islet
mone necessary for the development for growth antioxidant defence capacity was also impaired as
and differentiation of a tissue may have permanent evidenced by a reduced expression of antioxidant
structural consequences. There are several exam- enzymes manganese superoxide-dismutase, copper-
ples in rodent models of developmental program- zinc superoxide-dismutase, and heme oxygenase-1.
ming to suggest that the perinatal environment can Recent work by Theys and colleagues155 showed
have large and lasting effects on brain development. that male LP offspring demonstrated increases in
Neonatal treatment of rat pups during the second ROS production by islet mitochondria and this was
week of life with high levels of insulin induces per- associated with higher expression of mitochon-
manent changes in hypothalamic morphology, with drial subunits of the ETC NADH-ubiquinone
a reduced neuron density within the ventromedial oxireductase subunit 4L and peroxisome
hypothalamus,149 a region implicated in regulation proliferator-activated receptor-gamma and uncou-
of satiety. Structural alterations in this area of the pling protein-2 over-expression. It is suggested
brain could therefore contribute to the excess weight that this could explain the earlier development
gain and glucose intolerance in these rats neonatally of glucose intolerance in male LP offspring com-
treated with insulin. Leptin derangements during pared to females156 since this was only seen in
perinatal life have been implicated as an important males.
programming factor.150 The early neonatal plasma In the rat, uterine artery ligation induces de-
leptin surge has been very well characterized in both creased pyruvate oxidation in muscle tissue of
rats and mice; however, the functional significance young offspring, which leads to a chronic reduc-
of this surge is little known, with effects distinct from tion of ATP derived from oxidative phosphoryla-
those observed for adults in which leptin affects food tion. Glucose transporter 4 (GLUT4) recruitment,
intake and energy expenditure. Instead, neonatal glucose transport, and glycogen synthesis are com-
leptin has a neurotrophic role as evidenced by its promised, which contributes to insulin resistance
ability to stimulate neurite outgrowth from arcu- and hyperglycemia.157 Mitochondrial oxidative

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 87
Early life programming Fernandez-Twinn & Ozanne

phosphorylation was also found to be impaired in 4/4 Angelman syndrome (100%), 3/4 Prader-Willi
the liver158 and in fetal islet cells, mitochondrial syndrome (75%), 3/5 Down syndrome (60%), and
dysfunction leads to increased ROS production, re- 2/2 attention deficit hyperactivity disorder (100%)
duced ATP production and impaired mitochon- frontal cortex samples.167 One of the earliest re-
drial ETC complex I and III activities. This in turn ports that demonstrated methylation as mediat-
damages mitochondrial DNA leading to progressive ing the effects of early nutrition on adult disease
deterioration of beta cell function.159 The similar- susceptibility was provided by Waterland and Jir-
ity in mitochondrial dysfunction between the LP tle.168 More recently, Waterland and Jirtle showed
and uterine ligation models strongly suggests that that maternal obesity accumulated over successive
mitochondrial dysfunction plays a key role in the generations shifted the population distribution to-
metabolic phenotype of the offspring. ward increased adult body weight, and suggested
In a maternal diet-induced obesity model, mi- that epigenetic mechanisms are involved in this pro-
tochondrial complex II-III linked activity was re- cess.169 Spurred on by the concept that epigenet-
duced in skeletal muscle of male offspring.148 A re- ics would provide the fundamental mechanisms of
cent study showed that mouse oocytes and zygotes of early-life programming170 and therefore a means
dams exposed to diet-induced obesity demonstrated of intervention, many groups have invested in high-
increased mitochondrial potential, mitochondrial throughput methylation analyses. Global changes in
DNA content, and biogenesis,160 with evidence for DNA methylation have been observed in sheep that
raised ROS generation and oxidative stress, leading experienced alterations in vitamin B and methion-
to compromised mitochondrial metabolism very ine during the periconceptional period.171 There is
early in development. also evidence to suggest that the postnatal period is a
critical window in mammalian developmental epi-
Epigenetics of programming
genetics as demonstrated by developmental changes
Epigenetic mechanisms provide a tantalizing means in promoter methylation, which are associated with
of explaining how the early life environment may expression changes in liver tissue from embryonic
affect long-term chronic disease susceptibility. Epi- day 17.5 to postnatal day 21.172
genetic dysregulation is increasingly implicated in Maternal protein restriction has been shown to
cancer and many cancer types display hallmark epi- alter the methylation status of the promoters of the
genetic changes including histone modifications, GR,173 PPAR,174 and the angiotensin receptor,175
gene specific hypermethylation, and genome-wide which parallelled changes in gene expression. More
hypomethylation. Of these, global hypomethylation recent studies have shown that histone modifica-
is suggested as occurring early in transformation tions may also be influenced by the early environ-
and also important in tumor progression. The evi- ment. Both DNA methylation and histone acetyla-
dence for this comes from several studies showing tion changes in the PDX-1 promoter are observed
global hypomethylation in the adjacent healthy tis- in pancreatic beta cells of offspring of intrauter-
sue suggesting this change is an early event in the ine artery ligation pregnancies.176 These epigenetic
transformation process, resulting in karyotypic changes in the neonatal period were reversible by
instability and further oncogenic processes.161,162 histone deacetylase inhibition. However, after dia-
Controversially, several other investigations have betes onset, the PDX-1 locus was permanently si-
failed to detect reduced global methylcytosine lev- lenced through the methylation of the CpG island
els in benign lesions or during malignant tumor in the proximal promoter. Using a genome-wide
progression,163165 bringing into question the causal approach and whole islets isolated from male IUGR
nexus of genome-wide hypomethylation in carcino- rat offspring at 7 weeks of age (preceding diabetes),
genesis, and suggesting instead that it is a passive Thompson and colleagues found dysregulated cy-
inconsequential side effect of carcinogenesis.166 Hy- tosine methylation at 1400 loci, preferentially at
pomethylation has also been observed in neuro- conserved intergenic sequences, and frequently near
developmental disorders, for example, a reduction genes regulating islet vascularization, beta cell pro-
in methyl CpG binding protein 2 (MeCP2) expres- liferation, insulin secretion, and cell death. They
sion compared to age-matched controls was found also showed these to be associated with concor-
in 11/14 autism (79%), 9/9 Rett syndrome (100%), dant changes in mRNA expression thus representing

88 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

candidate loci for mediating the pathogenesis and and functions of epigenetic marks will continue to
progression of metabolic disease in later life.177 Al- puzzle for years to come.
terations in histone modifications are also impli-
Intervention strategies
cated in mediating the effect of caloric restriction
in the second trimester, with a programmed reduc- Nutritionists, clinicians, and researchers agree that
tion of GLUT4 expression in the offspring.178 Hu- optimal nutrition during early growth is vital to
man studies have also yielded evidence for the effect health outcome. Current advice and public health
of maternal diet on epigenetic marks in individ- initiatives promote healthy eating and these have
uals who were exposed to famine in-utero during reduced the incidence of low birth weight in most
the Dutch Hunger Winter. White blood cells taken developing countries. Obesity and higher levels of
from these individuals in their sixties, were found fat intake during pregnancy has emerged as a larger
to have less DNA methylation of the maternally im- problem in recent years and is potentially attributed
printed IGF2 gene compared with their same-sex with the rise in cases of high birth weight, correlat-
siblings, who were not exposed.179 The association ing with an increase in preeclampsia and obstetric
was specific for exposure during the periconcep- complications as well as an increased risk of fu-
tional period, reinforcing this period as a critical ture diabetes in the mother. It has vitally important
window for the establishment and maintenance of consequences for obesity and metabolic disease in
epigenetic marks. In a more recent study, cord blood children.
taken from IUGR neonates showed changes in cy- The need for therapeutic solutions to improve
tosine methylation, particularly in the hepatocyte glycemic control has seen the incretin Exendin-
nuclear factor 4alpha (HNF4A) gene, a well-known 4, a long lasting analogue glucagon-like peptide-1
diabetes candidate gene, and in other loci encoding emerge as an interesting candidate. Not only does it
HNF4A-interacting proteins.180 While both these improve insulin action through increasing beta cell
studies used peripheral blood, one main difference mass and function via induction of PDX-1 expres-
between these studies is the timing of insult. In the sion in beta cells,182 it also has effects on reducing
Heijman study, this was specific to the periconcep- appetite and food intake. In the restricted placental
tal period; this was not ascertainable in the Einstein growth model of IUGR, a 6-day neonatal course of
study. It should also be noted that individuals in the exendin-4 prevented the development of diabetes in
Dutch Hunger cohort were already insulin resistant the placentally restricted rat and normalised glucose
at the time of sampling, which could have affected tolerance after birth and into adulthood.183 beta cell
methylation status. mass and PDX-1 expression were also normalized
The role of methylation in complex chronic dis- and remarkably, this effect of neonatal exendin-4
eases, such as cardiovascular disease and obesity also restored the epigenetic state of the PDX-1 pro-
is beginning to emerge. One recent study demon- moter to control levels.184 The fetal rat pancreas,
strated cell-specific histone-methylation modi- however, unlike the human and sheep, does not de-
fications and expression of accompanying ly- velop insulin-containing cells until the second half
sine methyltransferases in the carotid arteries of gestation, and pancreatic remodeling with peaks
of apoE/+ mice born to hypercholesterolemic in replication and apoptosis occur in the early post-
apoE/ mothers.181 Furthermore, offspring from natal period at 12 weeks of age.185,186 Sheep, how-
apoE/ mothers demonstrated differences in hi- ever, as with humans, have nearly complete pancre-
stone triple-methylation modifications in vascular atic development before birth (reviewed By Gatford
endothelial and smooth muscle cells in response to a et al.187 ). Exendin-4 treatment in neonatal lambs
high cholesterol diet when compared with offspring from 1 day to 16 days after birth (1 nmol kg1 d1 )
from wild-type mothers. Thus, both in-utero pro- was able to increase glucose-stimulated insulin se-
gramming and postnatal hypercholesterolemia alter cretion by 155% in the twin IUGR lamb188 suggest-
epigenetic events in the vasculature. ing that this period of development is amenable to
All these studies suggest that critical windows in either or both of its effects that is, increasing beta
mammalian developmental epigenetics extend well cell mass and induction of PDX-1 expression. Such
beyond early embryonic development. The com- intervention may be useful in preventing future ad-
plicated interplay between the dynamics, positions, verse outcomes of the growth-restricted baby.

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 89
Early life programming Fernandez-Twinn & Ozanne

While recent animal studies suggest that mater- coronary heart disease: longitudinal study. BMJ 319: 1403
nal dietary supplementation with methyl donors 1407.
10. Caughey, R.W. & K.B. Michels. 2009. Birth weight and
and cofactors in late pregnancy can ameliorate the
childhood leukemia: a meta-analysis and review of the cur-
metabolic effects of growth restriction,187 we are still rent evidence. Int. J. Cancer 124: 26582670.
a long way from any kind of human intervention. 11. English, P.B., D.E. Goldberg, C. Wolff & D. Smith. 2003.
Parental and birth characteristics in relation to testicular
Summary cancer risk among males born between 1960 and 1995 in
California (United States). Cancer Causes Control 14: 815
Taking a global perspective of all the various ani-
825.
mal models of suboptimal early life, it is remark- 12. Kaijser, M., O. Akre, S. Cnattingius & A. Ekbom. 2005.
able that without exception, all of these mod- Preterm birth, low birth weight, and risk for esophageal
els, sooner or later, converge on an offspring adenocarcinoma. Gastroenterology 128: 607609.
phenotype of metabolic disease. Our understand- 13. Xu, X., A.B. Dailey, M. Peoples-Sheps, et al. 2009. Birth
weight as a risk factor for breast cancer: a meta-analysis
ing of disease mechanisms has been enhanced by
of 18 epidemiological studies. J. Womens Health (Larchmt)
each of these models, which meet in a crossroads 18: 11691178.
of metabolic dysregulation, with progression to as- 14. Xue, F. & K.B. Michels. 2007. Intrauterine factors and risk
sociated pathologies such as cardiovascular disease, of breast cancer: a systematic review and meta-analysis of
hypertension, and cancer radiating from this point. current evidence. Lancet Oncol. 8: 10881100.
15. Innes, K., T. Byers & M. Schymura. 2000. Birth character-
With recent technological advances, such as the de-
istics and subsequent risk for breast cancer in very young
velopment of deep sequencing, answers to some of women. Am. J. Epidemiol. 152: 11211128.
the remaining mechanistic questions should start to 16. Sanderson, M., M.A. Williams, K.E. Malone, et al. 1996.
emerge. Perinatal factors and risk of breast cancer. Epidemiology 7:
3437.
Conflicts of interest 17. Mellemkjaer, L., M.L. Olsen, H.T. Sorensen, et al. 2003.
Birth weight and risk of early-onset breast cancer (Den-
The authors declare no conflicts of interest. mark). Cancer Causes Control 14: 6164.
18. Kousta, E. 2006. Premature adrenarche leads to polycystic
References ovary syndrome? Long-term consequences. Ann. N.Y. Acad.
Sci. 1092: 148157.
1. Ford, E.S., W.H. Giles & W.H. Dietz. 2002. Prevalence of the 19. Main, K.M., R.B. Jensen, C. Asklund, et al. 2006. Low
metabolic syndrome among US adults: findings from the birth weight and male reproductive function. Horm. Res.
third National Health and Nutrition Examination Survey. 65(Suppl 3): 116122.
JAMA 287: 356359. 20. Vikse, B.E., L.M. Irgens, T. Leivestad, et al. 2008. Low birth
2. Diabetes-UK. Diabetes in the UK 2010: Key statistics on weight increases risk for end-stage renal disease. J. Am. Soc.
diabetes. 2010. Diabetes-UK, United Kingdom. Nephrol. 19: 151157.
3. Constancia, M., E. Angiolini, I. Sandovici, et al. 2005. Adap- 21. Luyckx, V.A. & B.M. Brenner. 2005. Low birth weight,
tation of nutrient supply to fetal demand in the mouse nephron number, and kidney disease. Kidney Int. Suppl.
involves interaction between the Igf2 gene and placental 68: S68S77.
transporter systems. Proc. Natl. Acad. Sci. USA 102: 19 219 22. Seidman, L.J., S.L. Buka, J.M. Goldstein, et al. 2000. The re-
19 224. lationship of prenatal and perinatal complications to cog-
4. Godfrey, K.M. & D.J. Barker. 2001. Fetal programming and nitive functioning at age 7 in the New England Cohorts
adult health. Public Health Nutr. 4: 611624. of the National Collaborative Perinatal Project. Schizophr.
5. Hales, C.N., D.J. Barker, P.M. Clark, et al. 1991. Fetal and Bull. 26: 309321.
infant growth and impaired glucose tolerance at age 64. 23. Nilsson, E., G. Stalberg, P. Lichtenstein, et al. 2005. Fetal
BMJ 303: 10191022. growth restriction and schizophrenia: a Swedish twin study.
6. Laitinen, J., K. Pietilainen, M. Wadsworth, et al. 2004. Pre- Twin Res. Hum. Genet. 8: 402408.
dictors of abdominal obesity among 31-yr-old men and 24. Lin, H.C., H.C. Lee, C.H. Tang & Y.H. Chen. 2010. The rela-
women born in Northern Finland in 1966. Eur. J. Clin. tion between maternal schizophrenia and low birth weight
Nutr. 58: 180190. is modified by paternal age. Can. J. Psychiatry 55: 377
7. Phillips, D.I. 1998. Birth weight and the future develop- 385.
ment of diabetes. A review of the evidence. Diabetes Care 25. Vestergaard, M., O. Basso, T.B. Henriksen, et al. 2002. Risk
21(Suppl 2): B150B155. factors for febrile convulsions. Epidemiology 13: 282287.
8. Eriksson, J.G., T. Forsen, J. Tuomilehto, et al. 1999. Catch- 26. Vestergaard, M. & J. Christensen. 2009. Register-based
up growth in childhood and death from coronary heart studies on febrile seizures in Denmark. Brain Dev. 31: 372
disease: longitudinal study. BMJ 318: 427431. 377.
9. Forsen, T., J.G. Eriksson, J. Tuomilehto, et al. 1999. Growth 27. Knip, M. & H.K. Akerblom. 2005. Early nutrition and later
in utero and during childhood among women who develop diabetes risk. Adv. Exp. Med. Biol. 569: 142150.

90 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

28. Yajnik, C.S. 2004. Early life origins of insulin resistance and 45. Singla, P.N., M. Tyagi, A. Kumar, et al. 1997. Fetal growth
type 2 diabetes in India and other Asian countries. J. Nutr. in maternal anaemia. J. Trop. Pediatr. 43: 8992.
134: 205210. 46. Steer, P., M.A. Alam, J. Wadsworth & A. Welch. 1995. Re-
29. Hattersley, A.T. & J.E. Tooke. 1999. The fetal insulin hy- lation between maternal haemoglobin concentration and
pothesis: an alternative explanation of the association of birth weight in different ethnic groups. BMJ 310: 489
low birthweight with diabetes and vascular disease. Lancet 491.
353: 17891792. 47. Allen, L.H. 2001. Biological mechanisms that might under-
30. Freathy, R.M., A.J. Bennett, S.M. Ring, et al. 2009. Type lie irons effects on fetal growth and preterm birth. J. Nutr.
2 diabetes risk alleles are associated with reduced size at 131(2S-2): 581S589S.
birth. Diabetes 58: 14281433. 48. Rosario, J.F., M.P. Gomez & P. Anbu. 2008. Does the ma-
31. Dupuis, J., C. Langenberg, I. Prokopenko, et al. 2010. New ternal micronutrient deficiency (copper or zinc or vitamin
genetic loci implicated in fasting glucose homeostasis and E) modulate the expression of placental 11 beta hydrox-
their impact on type 2 diabetes risk. Nat. Genet. 42: 105 ysteroid dehydrogenase-2 per se predispose offspring to
116. insulin resistance and hypertension in later life? Indian J.
32. Hales, C.N. & D.J. Barker. 2001. The thrifty phenotype Physiol. Pharmacol. 52: 355365.
hypothesis. Br. Med. Bull. 60: 520. 49. Christian, P., C.P. Stewart, S.C. LeClerq, et al. 2009. Ante-
33. Ravelli, A.C., J.H. Van Der Meulen, R.P. Michels, et al. natal and postnatal iron supplementation and childhood
1998. Glucose tolerance in adults after prenatal exposure mortality in rural Nepal: a prospective follow-up in a ran-
to famine. Lancet 351: 173177. domized, controlled community trial. Am. J. Epidemiol.
34. Bo, S., P. Cavallo-Perin, L. Scaglione, et al. 2000. Low birth- 170: 11271136.
weight and metabolic abnormalities in twins with increased 50. Fall, C.H., D.J. Fisher, C. Osmond & B.M. Margetts. 2009.
susceptibility to Type 2 diabetes mellitus. Diabet. Med. 17: Multiple micronutrient supplementation during preg-
365370. nancy in low-income countries: a meta-analysis of effects
35. Grunnet, L., S. Vielwerth, A. Vaag & P. Poulsen. 2007. Birth on birth size and length of gestation. Food Nutr. Bull. 30(4
weight is nongenetically associated with glucose intolerance Suppl): S533S546.
in elderly twins, independent of adult obesity. J. Intern. 51. Reinisch, J.M., N.G. Simon, W.G. Karow & R. Gandelman.
Med. 262: 96103. 1978. Prenatal exposure to prednisone in humans and an-
36. Poulsen, P., K.O. Kyvik, A. Vaag & H. Beck-Nielsen. 1999. imals retards intrauterine growth. Science 202: 436438.
Heritability of type II (non-insulin-dependent) diabetes 52. Clark, P.M. 1998. Programming of the hypothalamo-
mellitus and abnormal glucose tolerancea population- pituitary-adrenal axis and the fetal origins of adult disease
based twin study. Diabetologia 42: 139145. hypothesis. Eur. J. Pediatr. 157(Suppl 1): S7S10.
37. Dunger, D.B., B. Salgin & K.K. Ong. 2007. Session 7: early 53. Vallee, M., W. Mayo, S. Maccari, et al. 1996. Long-term
nutrition and later health early developmental pathways of effects of prenatal stress and handling on metabolic pa-
obesity and diabetes risk. Proc. Nutr. Soc. 66: 451457. rameters: relationship to corticosterone secretion response.
38. Vaag, A., C.B. Jensen, P. Poulsen, et al. 2006. Metabolic Brain Res. 712: 287292.
aspects of insulin resistance in individuals born small for 54. Lesage, J., F. Del-Favero, M. Leonhardt, et al. 2004. Prena-
gestational age. Horm. Res. 65(Suppl 3): 137143. tal stress induces intrauterine growth restriction and pro-
39. Gluckman, P.D. & M.A. Hanson. 2004. Maternal constraint grammes glucose intolerance and feeding behaviour dis-
of fetal growth and its consequences. Semin. Fetal Neonatal. turbances in the aged rat. J. Endocrinol. 181: 291296.
Med. 9: 419425. 55. Pepe, G.J., M.G. Burch & E.D. Albrecht. 2001. Localization
40. Jablonka, E. & G. Raz. 2009. Transgenerational epigenetic and developmental regulation of 11beta-hydroxysteroid
inheritance: prevalence, mechanisms, and implications for dehydrogenase-1 and -2 in the baboon syncytiotro-
the study of heredity and evolution. Q. Rev. Biol. 84: 131 phoblast. Endocrinology 142: 6880.
176. 56. Seckl, J.R., M. Cleasby & M.J. Nyirenda. 2000. Glucocorti-
41. Giussani, D.A., P.S. Phillips, S. Anstee & D.J. Barker. 2001. coids, 11beta-hydroxysteroid dehydrogenase, and fetal pro-
Effects of altitude versus economic status on birth weight gramming. Kidney Int. 57: 14121417.
and body shape at birth. Pediatr. Res. 49: 490494. 57. McMullen, S., J.C. Osgerby, L.M. Thurston, et al. 2004.
42. Khalid, M.E., M.E. Ali & K.Z. Ali. 1997. Full-term birth Alterations in placental 11 beta-hydroxysteroid dehydro-
weight and placental morphology at high and low altitude. genase (11 betaHSD) activities and fetal cortisol:cortisone
Int. J. Gynaecol. Obstet. 57: 259265. ratios induced by nutritional restriction prior to concep-
43. Julian, C.G., M.J. Wilson, M. Lopez, et al. 2009. Augmented tion and at defined stages of gestation in ewes. Reproduction
uterine artery blood flow and oxygen delivery protect An- 127: 717725.
deans from altitude-associated reductions in fetal growth. 58. Challis, J.R., D.M. Sloboda, N. Alfaidy, et al. 2002.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 296: R1564 Prostaglandins and mechanisms of preterm birth. Repro-
R1575. duction 124: 117.
44. McLean, E., M. Cogswell, I. Egli, et al. 2009. Worldwide 59. Phillips, D.I., D.J. Barker, C.H. Fall, et al. 1998. Elevated
prevalence of anaemia, WHO Vitamin and Mineral Nutri- plasma cortisol concentrations: a link between low birth
tion Information System, 19932005. Public Health Nutr. weight and the insulin resistance syndrome? J. Clin. En-
12: 444454. docrinol. Metab. 83: 757760.

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 91
Early life programming Fernandez-Twinn & Ozanne

60. Crowther, N.J., N. Cameron, J. Trusler & I.P. Gray. 1998. long-term implications of maternal obesity on the mother
Association between poor glucose tolerance and rapid post and her offspring. BJOG 113: 11261133.
natal weight gain in seven-year-old children. Diabetologia 77. Stewart, F.M., D.J. Freeman, J.E. Ramsay, et al. 2007. Lon-
41: 11631167. gitudinal assessment of maternal endothelial function and
61. Forsen, T., J. Eriksson, J. Tuomilehto, et al. 2000. The fe- markers of inflammation and placental function through-
tal and childhood growth of persons who develop type 2 out pregnancy in lean and obese mothers. J. Clin. En-
diabetes. Ann. Intern. Med. 133: 176182. docrinol. Metab. 92: 969975.
62. Yajnik, C. 2000. Interactions of perturbations in intrauter- 78. Ramsay, J.E., W.R. Ferrell, L. Crawford, et al. 2002. Mater-
ine growth and growth during childhood on the risk of nal obesity is associated with dysregulation of metabolic,
adult-onset disease. Proc. Nutr. Soc. 59: 257265. vascular, and inflammatory pathways. J. Clin. Endocrinol.
63. Crowther, N.J., J. Trusler, N. Cameron, et al. 2000. Relation Metab. 87: 42314237.
between weight gain and beta-cell secretory activity and 79. Toescu, V., S.L. Nuttall, U. Martin, et al. 2002. Oxidative
non-esterified fatty acid production in 7-year-old African stress and normal pregnancy. Clin. Endocrinol. (Oxf.) 57:
children: results from the Birth to Ten study. Diabetologia 609613.
43: 978985. 80. Sanchez-Vera, I., B. Bonet, M. Viana, et al. 2007. Changes
64. Lanigan, J. & A. Singhal. 2009. Early nutrition and long- in plasma lipids and increased low-density lipoprotein sus-
term health: a practical approach. Proc. Nutr. Soc. 68: 422 ceptibility to oxidation in pregnancies complicated by ges-
429. tational diabetes: consequences of obesity. Metabolism 56:
65. Chan, K.K., L.F. Ho & T.T. Lao. 2003. Nutritional intake and 15271533.
placental size in gestational diabetic pregnanciesa prelim- 81. Zwirska-Korczala, K., J. Jagodzinska, T. Wielkoszynski, et al.
inary observation. Placenta 24: 985988. 2001. [Plasma oxysterols and vitamin E concentrations and
66. Evers, I.M., H.W. de Valk, B.W. Mol, et al. 2002. Macro- lipid profile in morbidly obese women]. Pol. Arch. Med.
somia despite good glycaemic control in Type I diabetic Wewn. 106: 909915.
pregnancy; results of a nationwide study in The Nether- 82. Alkazemi, D., G. Egeland, J. Vaya, et al. 2008. Oxysterol as a
lands. Diabetologia 45: 14841489. marker of atherogenic dyslipidemia in adolescence. J. Clin.
67. Savona-Ventura, C. & M. Chircop. 2003. Birth weight influ- Endocrinol. Metab. 93: 42824289.
ence on the subsequent development of gestational diabetes 83. Jansson, T., L. Myatt & T.L. Powell. 2009. The role of tro-
mellitus. Acta Diabetol. 40: 101104. phoblast nutrient and ion transporters in the development
68. Pettitt, D.J., K.M. Narayan, R.L. Hanson & W.C. Knowler. of pregnancy complications and adult disease. Curr. Vasc.
1996. Incidence of diabetes mellitus in women following Pharmacol. 7: 521533.
impaired glucose tolerance in pregnancy is lower than fol- 84. Fowden, A.L., J.W. Ward, F.P. Wooding, et al. 2006. Pro-
lowing impaired glucose tolerance in the non-pregnant gramming placental nutrient transport capacity. J. Physiol.
state. Diabetologia 39: 13341337. 572(Pt 1): 515.
69. Williams, M.A., I. Emanuel, C. Kimpo, et al. 1999. A 85. Myatt, L. 2006. Placental adaptive responses and fetal pro-
population-based cohort study of the relation between ma- gramming. J. Physiol. 572(Pt 1): 2530.
ternal birthweight and risk of gestational diabetes mellitus 86. Jansson, N., J. Pettersson, A. Haafiz, et al. 2006. Down-
in four racial/ethnic groups. Paediatr. Perinat. Epidemiol. regulation of placental transport of amino acids precedes
13: 452465. the development of intrauterine growth restriction in rats
70. Nelson, S.M., P. Matthews & L. Poston. 2010. Maternal fed a low protein diet. J. Physiol. 576(Pt 3): 935946.
metabolism and obesity: modifiable determinants of preg- 87. Fowden, A.L., A.N. Sferruzzi-Perri, P.M. Coan, et al. 2009.
nancy outcome. Hum. Reprod. Update 16: 255275. Placental efficiency and adaptation: endocrine regulation.
71. Catalano, P.M., L. Presley, J. Minium & S. Hauguel-de J. Physiol. 587(Pt 14): 34593472.
Mouzon. 2009. Fetuses of obese mothers develop insulin 88. Woodall, S.M., B.H. Breier, B.M. Johnston & P.D. Gluck-
resistance in utero. Diabetes Care 32: 10761080. man. 1996. A model of intrauterine growth retardation
72. Voldner, N., K.F. Froslie, K. Bo, et al. 2008. Modifiable caused by chronic maternal undernutrition in the rat:
determinants of fetal macrosomia: role of lifestyle-related effects on the somatotrophic axis and postnatal growth.
factors. Acta Obstet. Gynecol. Scand. 87: 423429. J. Endocrinol. 150: 231242.
73. Olsen, S.F., T.I. Halldorsson, W.C. Willett, et al. 2007. 89. Coan, P.M., Vaughan, O.R., Sekita, Y., et al. 2010. Adapta-
Milk consumption during pregnancy is associated with in- tions in placental phenotype support fetal growth during
creased infant size at birth: prospective cohort study. Am. undernutrition of pregnant mice. J. Physiol. 588(Pt 3): 527
J. Clin. Nutr. 86: 11041110. 538.
74. Hedderson, M.M., M.A. Williams, V.L. Holt, et al. 2008. 90. Langley-Evans, S.C., D.S. Gardner & A.A. Jackson. 1996.
Body mass index and weight gain prior to pregnancy and Association of disproportionate growth of fetal rats in late
risk of gestational diabetes mellitus. Am. J. Obstet. Gynecol. gestation with raised systolic blood pressure in later life.
198: 409 e1e7. J. Reprod. Fertil. 106: 307312.
75. Huda, S.S., L.E. Brodie & N. Sattar. 2009. Obesity in preg- 91. Fernandez-Twinn, D.S., S.E. Ozanne, S. Ekizoglou, et al.
nancy: prevalence and metabolic consequences. Semin. Fe- 2003. The maternal endocrine environment in the low-
tal Neonatal. Med. 15: 7076. protein model of intra-uterine growth restriction. Br. J.
76. Catalano, P.M. & H.M. Ehrenberg. 2006. The short- and Nutr. 90: 815822.

92 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

92. Hay, W.W. Jr. 1994. Placental transport of nutrients to the 108. Vickers, M.H., P.D. Gluckman, A.H. Coveny, et al. 2005.
fetus. Horm. Res. 42: 215222. Neonatal leptin treatment reverses developmental pro-
93. Sugden, M.C., M.L. Langdown, M.J. Munns & M.J. Holness. gramming. Endocrinology 146: 42114216.
2001. Maternal glucocorticoid treatment modulates pla- 109. Dumortier, O., B. Blondeau, B. Duvillie, et al. 2007.
cental leptin and leptin receptor expression and materno- Different mechanisms operating during different critical
fetal leptin physiology during late pregnancy, and elicits hy- time-windows reduce rat fetal beta cell mass due to a mater-
pertension associated with hyperleptinaemia in the early- nal low-protein or low-energy diet. Diabetologia 50: 2495
growth-retarded adult offspring. Eur. J. Endocrinol. 145: 2503.
529539. 110. Snoeck, A., C. Remacle, B. Reusens & J.J. Hoet. 1990. Effect
94. Lesage, J., D. Hahn, M. Leonhardt, et al. 2002. Maternal of a low protein diet during pregnancy on the fetal rat
undernutrition during late gestation-induced intrauterine endocrine pancreas. Biol. Neonate. 57: 107118.
growth restriction in the rat is associated with impaired 111. Langley-Evans, S.C. & M. Nwagwu. 1998. Impaired growth
placental GLUT3 expression, but does not correlate with and increased glucocorticoid-sensitive enzyme activities in
endogenous corticosterone levels. J. Endocrinol. 174: 37 tissues of rat fetuses exposed to maternal low protein diets.
43. Life Sci. 63: 605615.
95. Efstratiadis, A. 1998. Genetics of mouse growth. Int. J. Dev. 112. Hoet, J.J., S. Dahri, A. Snoeck, et al. 1992. [Importance
Biol. 42: 955976. of diets and their effect on fetal development: function
96. Martin, P.M. & A.E. Sutherland. 2001. Exogenous amino and structure of the endocrine pancreas following protein
acids regulate trophectoderm differentiation in the mouse deficiency during intrauterine life]. Bull. Mem. Acad. R.
blastocyst through an mTOR-dependent pathway. Dev. Med. Belg. 147: 174181; discussion 8183.
Biol. 240: 182193. 113. Petry, C.J., M.W. Dorling, D.B. Pawlak, et al. 2001. Diabetes
97. Wen, H.Y., S. Abbasi, R.E. Kellems & Y. Xia. 2005. mTOR: in old male offspring of rat dams fed a reduced protein diet.
a placental growth signaling sensor. Placenta 26(Suppl A): Int. J. Exp. Diabetes Res. 2: 139143.
S63S69. 114. Ozanne, S.E., B.T. Nave, C.L. Wang, et al. 1997. Poor fetal
98. Roos, S., N. Jansson, I. Palmberg, et al. 2007. Mammalian nutrition causes long-term changes in expression of insulin
target of rapamycin in the human placenta regulates leucine signaling components in adipocytes. Am. J. Physiol. 273(1
transport and is down-regulated in restricted fetal growth. Pt 1): E46E51.
J. Physiol. 582(Pt 1): 449459. 115. Ozanne, S.E., G.S. Olsen, L.L. Hansen, et al. 2003. Early
99. Palm, M., O. Axelsson, L. Wernroth & S. Basu. 2009. growth restriction leads to down regulation of protein
F(2)-isoprostanes, tocopherols and normal pregnancy. Free kinase C zeta and insulin resistance in skeletal muscle.
Radic. Res. 43: 546552. J. Endocrinol. 177: 235241.
100. Toescu, V., S.L. Nuttall, U. Martin, et al. 2004. Changes 116. Ozanne, S.E., C.B. Jensen, K.J. Tingey, et al. 2006. Decreased
in plasma lipids and markers of oxidative stress in normal protein levels of key insulin signalling molecules in adipose
pregnancy and pregnancies complicated by diabetes. Clin. tissue from young men with a low birthweight: potential
Sci. (Lond.) 106: 9398. link to increased risk of diabetes? Diabetologia 49: 2993
101. Burton, G.J., J. Hempstock & E. Jauniaux. 2001. Nutrition 2999.
of the human fetus during the first trimestera review. 117. Ozanne, S.E., C.B. Jensen, K.J. Tingey, et al. 2005. Low
Placenta 22(Suppl A): S70S77. birthweight is associated with specific changes in muscle
102. Myatt L. 2010. Review: reactive oxygen and nitrogen insulin-signalling protein expression. Diabetologia 48: 547
species and functional adaptation of the placenta. Placenta 552.
31(Suppl): S66S69. 118. Hales, C.N., M. Desai, S.E. Ozanne & N.J. Crowther. 1996.
103. Symonds, M.E. & H. Budge. 2009. Nutritional models of the Fishing in the stream of diabetes: from measuring insulin
developmental programming of adult health and disease. to the control of fetal organogenesis. Biochem. Soc. Trans.
Proc. Nutr. Soc. 68: 173178. 24: 341350.
104. Garofano, A., P. Czernichow & B. Breant. 1997. In utero un- 119. Jennings, B.J., S.E. Ozanne, M.W. Dorling & C.N. Hales.
dernutrition impairs rat beta-cell development. Diabetolo- 1999. Early growth determines longevity in male rats and
gia 40: 12311234. may be related to telomere shortening in the kidney. FEBS
105. Garofano, A., P. Czernichow & B. Breant. 1998. Beta- Lett. 448: 48.
cell mass and proliferation following late fetal and early 120. Tarry-Adkins, J.L., J.H. Chen, N.S. Smith, et al. 2009.
postnatal malnutrition in the rat. Diabetologia 41: 1114 Poor maternal nutrition followed by accelerated postnatal
1120. growth leads to telomere shortening and increased markers
106. Vickers, M.H., B.H. Breier, W.S. Cutfield, et al. 2000. Fe- of cell senescence in rat islets. FASEB J. 23: 15211528.
tal origins of hyperphagia, obesity, and hypertension and 121. Ozanne, S.E. & C.N. Hales. 2004. Lifespan: catch-up growth
postnatal amplification by hypercaloric nutrition. Am. J. and obesity in male mice. Nature 427: 411412.
Physiol. Endocrinol. Metab. 279: E83E87. 122. Ozanne, S.E. & C. Nicholas Hales. 2005. Poor fetal growth
107. Vickers, M.H., B.A. Ikenasio & B.H. Breier. 2001. IGF-I followed by rapid postnatal catch-up growth leads to pre-
treatment reduces hyperphagia, obesity, and hypertension mature death. Mech. Ageing Dev. 126: 852854.
in metabolic disorders induced by fetal programming. En- 123. Ozanne, S.E., R. Lewis, B.J. Jennings & C.N. Hales. 2004.
docrinology 142: 39643973. Early programming of weight gain in mice prevents the

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 93
Early life programming Fernandez-Twinn & Ozanne

induction of obesity by a highly palatable diet. Clin. Sci. diovascular and metabolic physiology. Am. J. Physiol. En-
(Lond.) 106: 141145. docrinol. Metab. 287: E863E870.
124. Isganaitis, E., J. Jimenez-Chillaron, M. Woo, et al. 2009. Ac- 140. Lindsay, R.S., R.M. Lindsay, C.R. Edwards & J.R. Seckl.
celerated postnatal growth increases lipogenic gene expres- 1996. Inhibition of 11-beta-hydroxysteroid dehydrogenase
sion and adipocyte size in low-birth weight mice. Diabetes in pregnant rats and the programming of blood pressure
58: 11921200. in the offspring. Hypertension 27: 12001204.
125. Bol, V.V., A.I. Delattre, B. Reusens, et al. 2009. Forced 141. Seckl, J.R. 2004. Prenatal glucocorticoids and long-term
catch-up growth after fetal protein restriction alters the programming. Eur. J. Endocrinol. 151(Suppl 3): U49U62.
adipose tissue gene expression program leading to obesity 142. McMullen, S. & S.C. Langley-Evans. 2005. Sex-specific ef-
in adult mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. fects of prenatal low-protein and carbenoxolone exposure
297: R291R299. on renal angiotensin receptor expression in rats. Hyperten-
126. Sutton, G.M., A.V. Centanni & A.A. Butler. 2010. Protein sion 46: 13741380.
malnutrition during pregnancy in C57BL/6J mice results in 143. Holemans, K., L. Aerts & F.A. Van Assche. 2003. Fetal
offspring with altered circadian physiology before obesity. growth restriction and consequences for the offspring in
Endocrinology 151: 15701580. animal models. J. Soc. Gynecol. Investig. 10: 392399.
127. Robinson, L.K. & L.S. Hurley. 1981. Effect of maternal 144. Van Assche, F.A., K. Holemans & L. Aerts. 1998. Fetal
zinc deficiency of food restriction on rat fetal pancreas. 2. growth and consequences for later life. J. Perinat. Med.
Insulin and glucagon. J. Nutr. 111: 869877. 26: 337346.
128. Padmavathi, I.J., Y.D. Kishore, L. Venu, et al. 2009. Prenatal 145. Samuelsson, A.M., P.A. Matthews, M. Argenton, et al. 2008.
and perinatal zinc restriction: effects on body composition, Diet-induced obesity in female mice leads to offspring hy-
glucose tolerance and insulin response in rat offspring. Exp. perphagia, adiposity, hypertension, and insulin resistance:
Physiol. 94: 761769. a novel murine model of developmental programming.
129. Lisle, S.J., R.M. Lewis, C.J. Petry, et al. 2003. Effect of mater- Hypertension 51: 383392.
nal iron restriction during pregnancy on renal morphology 146. Kirk, S.L., A.M. Samuelsson, M. Argenton, et al. 2009. Ma-
in the adult rat offspring. Br. J. Nutr. 90: 3339. ternal obesity induced by diet in rats permanently influ-
130. Lewis, R.M., A.J. Forhead, C.J. Petry, et al. 2002. Long-term ences central processes regulating food intake in offspring.
programming of blood pressure by maternal dietary iron PLoS One 4: e5870; 113.
restriction in the rat. Br. J. Nutr. 88: 283290. 147. Oben, J.A., A. Mouralidarane, A.M. Samuelsson, et al. 2010.
131. Illsley, N.P., I. Caniggia & S. Zamudio. 2010. Placen- Maternal obesity during pregnancy and lactation programs
tal metabolic reprogramming: do changes in the mix of the development of offspring non-alcoholic fatty liver dis-
energy-generating substrates modulate fetal growth? Int. J. ease in mice. J. Hepatol. 52: 913920.
Dev. Biol. 54: 409419. 148. Shelley, P., M.S. Martin-Gronert, A. Rowlerson, et al. 2009.
132. Schaffer, L., J. Vogel, C. Breymann, et al. 2006. Preserved Altered skeletal muscle insulin signaling and mitochon-
placental oxygenation and development during severe sys- drial complex II-III linked activity in adult offspring of
temic hypoxia. Am. J. Physiol. Regul. Integr. Comp. Physiol. obese mice. Am. J. Physiol. Regul. Integr. Comp. Physiol.
290: R844R851. 297: R675R681.
133. Haggarty, P., S. Allstaff, G. Hoad, et al. 2002. Placental 149. Harder, T., A. Plagemann, W. Rohde & G. Dorner. 1998.
nutrient transfer capacity and fetal growth. Placenta 23: Syndrome X-like alterations in adult female rats due to
8692. neonatal insulin treatment. Metabolism 47: 855862.
134. Styrud, J., U.J. Eriksson, V. Grill & I. Swenne. 2005. Exper- 150. Yura, S., H. Itoh, N. Sagawa, et al. 2005. Role of premature
imental intrauterine growth retardation in the rat causes leptin surge in obesity resulting from intrauterine under-
a reduction of pancreatic B-cell mass, which persists into nutrition. Cell Metab. 1: 371378.
adulthood. Biol. Neonate. 88: 122128. 151. Bouret, S.G., S.J. Draper & R.B. Simerly. 2004. Formation
135. Simmons, R.A., L.J. Templeton & S.J. Gertz. 2001. In- of projection pathways from the arcuate nucleus of the
trauterine growth retardation leads to the development of hypothalamus to hypothalamic regions implicated in the
type 2 diabetes in the rat. Diabetes 50: 22792286. neural control of feeding behavior in mice. J. Neurosci. 24:
136. Merlet-Benichou, C., T. Gilbert, M. Muffat-Joly, et al. 27972805.
1994. Intrauterine growth retardation leads to a permanent 152. Turrens, J.F. 2003. Mitochondrial formation of reactive oxy-
nephron deficit in the rat. Pediatr. Nephrol. 8: 175180. gen species. J Physiol 552(Pt 2): 335344.
137. Schreuder, M.F., J.R. Nyengaard, M. Fodor, et al. 2005. 153. Passos, J.F., G. Saretzki, S. Ahmed, et al. 2007. Mitochon-
Glomerular number and function are influenced by spon- drial dysfunction accounts for the stochastic heterogene-
taneous and induced low birth weight in rats. J. Am. Soc. ity in telomere-dependent senescence. PLoS Biol. 5: 1138
Nephrol. 16: 29132919. 1151.
138. Boloker, J., S.J. Gertz & R.A. Simmons. 2002. Gestational 154. Tarry-Adkins, J.L., J.H. Chen, R.H. Jones, et al. 2010. Poor
diabetes leads to the development of diabetes in adulthood maternal nutrition leads to alterations in oxidative stress,
in the rat. Diabetes 51: 14991506. antioxidant defense capacity, and markers of fibrosis in rat
139. ORegan, D., C.J. Kenyon, J.R. Seckl & M.C. Holmes. 2004. islets: potential underlying mechanisms for development
Glucocorticoid exposure in late gestation in the rat perma- of the diabetic phenotype in later life. FASEB J. 24: 2762
nently programs gender-specific differences in adult car- 2771.

94 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.
Fernandez-Twinn & Ozanne Early life programming

155. Theys, N., T. Bouckenooghe, M.T. Ahn, et al. 2009. Ma- 170. Ozanne, S.E. & M. Constancia. 2007. Mechanisms of dis-
ternal low-protein diet alters pancreatic islet mitochon- ease: the developmental origins of disease and the role of
drial function in a sex-specific manner in the adult rat. the epigenotype. Nat. Clin. Pract. Endocrinol. Metab. 3:
Am. J. Physiol. Regul. Integr. Comp. Physiol. 297: R1516 539546.
R1525. 171. Sinclair, K.D., C. Allegrucci, R. Singh, et al. 2007. DNA
156. Fernandez-Twinn, D.S., A. Wayman, S. Ekizoglou, et al. methylation, insulin resistance, and blood pressure in off-
2005. Maternal protein restriction leads to hyperinsuline- spring determined by maternal periconceptional B vita-
mia and reduced insulin-signaling protein expression in min and methionine status. Proc. Natl. Acad. Sci. USA 104:
21-mo-old female rat offspring. Am. J. Physiol. Regul. In- 1935119356.
tegr. Comp. Physiol. 288: R368R373. 172. Waterland, R.A., R. Kellermayer, M.T. Rached, et al. 2009.
157. Selak, M.A., B.T. Storey, I. Peterside & R.A. Simmons. Epigenomic profiling indicates a role for DNA methylation
2003. Impaired oxidative phosphorylation in skeletal mus- in early postnatal liver development. Hum. Mol. Genet. 18:
cle of intrauterine growth-retarded rats. Am. J. Physiol. En- 30263038.
docrinol. Metab. 285: E130E137. 173. Lillycrop, K.A., E.S. Phillips, A.A. Jackson, et al. 2005. Di-
158. Peterside, I.E., M.A. Selak & R.A. Simmons. 2003. Im- etary protein restriction of pregnant rats induces and folic
paired oxidative phosphorylation in hepatic mitochondria acid supplementation prevents epigenetic modification of
in growth-retarded rats. Am. J. Physiol. Endocrinol. Metab. hepatic gene expression in the offspring. J. Nutr. 135: 1382
285: E1258E1266. 1386.
159. Simmons, R.A., I. Suponitsky-Kroyter & M.A. Selak. 2005. 174. Lillycrop, K.A., E.S. Phillips, C. Torrens, et al. 2008. Feeding
Progressive accumulation of mitochondrial DNA muta- pregnant rats a protein-restricted diet persistently alters the
tions and decline in mitochondrial function lead to beta- methylation of specific cytosines in the hepatic PPAR alpha
cell failure. J. Biol. Chem. 280: 2878528791. promoter of the offspring. Br. J. Nutr. 100: 278282.
160. Igosheva, N., A.Y. Abramov, L. Poston, et al. 2010. Mater- 175. Bogdarina, I., S. Welham, P.J. King, et al. 2007. Epigenetic
nal diet-induced obesity alters mitochondrial activity and modification of the renin-angiotensin system in the fetal
redox status in mouse oocytes and zygotes. PLoS One 5: programming of hypertension. Circ. Res. 100: 520526.
e10074;18. 176. Park, J.H., D.A. Stoffers, R.D. Nicholls & R.A. Simmons.
161. Goelz, S.E., B. Vogelstein, S.R. Hamilton & A.P. Feinberg. 2008. Development of type 2 diabetes following intrauter-
1985. Hypomethylation of DNA from benign and malig- ine growth retardation in rats is associated with progressive
nant human colon neoplasms. Science 228: 187190. epigenetic silencing of Pdx1. J. Clin. Invest. 118: 23162324.
162. Suter, C.M., D.I. Martin & R.L. Ward. 2004. Hypomethy- 177. Thompson, R.F., M.J. Fazzari, H. Niu, et al. 2010. Experi-
lation of L1 retrotransposons in colorectal cancer and ad- mental intrauterine growth restriction induces alterations
jacent normal tissue. Int. J. Colorectal. Dis. 19: 95101. in DNA methylation and gene expression in pancreatic
163. Yegnasubramanian, S., M.C. Haffner, Y. Zhang, et al. islets of rats. J. Biol. Chem. 285: 1511115118.
2008. DNA hypomethylation arises later in prostate cancer 178. Raychaudhuri, N., S. Raychaudhuri, M. Thamotharan
progression than CpG island hypermethylation and con- & S.U. Devaskar. 2008. Histone code modifications re-
tributes to metastatic tumor heterogeneity. Cancer Res. 68: press glucose transporter 4 expression in the intrauter-
89548967. ine growth-restricted offspring. J. Biol. Chem. 283: 13611
164. Florl, A.R., C. Steinhoff, M. Muller, et al. 2004. Coordinate 13626.
hypermethylation at specific genes in prostate carcinoma 179. Heijmans, B.T., E.W. Tobi, A.D. Stein, et al. 2008. Persistent
precedes LINE-1 hypomethylation. Br. J. Cancer 91: 985 epigenetic differences associated with prenatal exposure to
994. famine in humans. Proc. Natl. Acad. Sci. USA 105: 17046
165. Ehrlich, M., C.B. Woods, M.C. Yu, et al. 2006. Quantitative 17049.
analysis of associations between DNA hypermethylation, 180. Einstein, F., R.F. Thompson, T.D. Bhagat, et al. 2010. Cy-
hypomethylation, and DNMT RNA levels in ovarian tu- tosine methylation dysregulation in neonates following in-
mors. Oncogene 25: 26362645. trauterine growth restriction. PLoS One 5: e8887;111.
166. Wild, L. & J.M. Flanagan. 2010. Genome-wide hypomethy- 181. Alkemade, F.E., P. van Vliet, P. Henneman, et al. 2010.
lation in cancer may be a passive consequence of transfor- Prenatal exposure to apoE deficiency and postnatal hy-
mation. Biochim. Biophys. Acta. 1806: 5057. percholesterolemia are associated with altered cell-specific
167. Nagarajan, R.P., A.R. Hogart, Y. Gwye, et al. 2006. Reduced lysine methyltransferase and histone methylation patterns
MeCP2 expression is frequent in autism frontal cortex and in the vasculature. Am. J. Pathol. 176: 542548.
correlates with aberrant MECP2 promoter methylation. 182. Doyle, M.E. & J.M. Egan. 2007. Mechanisms of action of
Epigenetics 1: e1e11. glucagon-like peptide 1 in the pancreas. Pharmacol. Ther.
168. Waterland, R.A. & R.L. Jirtle. 2004. Early nutrition, epi- 113: 546593.
genetic changes at transposons and imprinted genes, and 183. Stoffers, D.A., B.M. Desai, D.D. DeLeon & R.A. Simmons.
enhanced susceptibility to adult chronic diseases. Nutrition 2003. Neonatal exendin-4 prevents the development of di-
20: 6368. abetes in the intrauterine growth retarded rat. Diabetes 52:
169. Waterland, R.A. 2008. Epigenetic epidemiology of obesity: 734740.
application of epigenomic technology. Nutr. Rev. 66(Suppl 184. Pinney, S.E., H. Niu, F. Li & R.A. Simmons. 2007.
1): S21S23. Neonatal exendin-4 administration normalises epigenetic

Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences. 95
Early life programming Fernandez-Twinn & Ozanne

modifications of the proximal promoter of Pdx-1. Early 187. Gatford, K.L., R.A. Simmons, M.J. De Blasio, et al. 2010.
Hum. Dev. 83(Suppl. 1): S71. Abstract 5B-4. Review: placental programming of postnatal diabetes and
185. Reddy, S. & R.B. Elliott. 1988. Ontogenic development of impaired insulin action after IUGR. Placenta 31(Suppl):
peptide hormones in the mammalian fetal pancreas. Expe- S60S65.
rientia 44: 19. 188. Gatford, K.L., S.A. Sulaiman, S.N.B. Mohammad, et al.
186. Scaglia, L., F.E. Smith & S. Bonner-Weir. 1995. Apoptosis 2009. Interventions to Prevent Diabetes after IUGR Prelim-
contributes to the involution of beta cell mass in the post inary Outcomes in the Twin IUGR Lamb. Fetal and Neonatal
partum rat pancreas. Endocrinology 136: 54615468. Physiology Workshop. Darwin. Australia.

96 Ann. N.Y. Acad. Sci. 1212 (2010) 7896 


c 2010 New York Academy of Sciences.

Vous aimerez peut-être aussi