You are on page 1of 13

REPRODUCTION

REVIEW

Sex differences in developmental programming models


Catherine E Aiken1,2 and Susan E Ozanne2
1
Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR Cambridge Comprehensive Biomedical
Research Centre, University of Cambridge, PO Box 223, Cambridge CB2 0SW, UK and 2Metabolic Research
Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ,
UK
Correspondence should be addressed to C E Aiken at Department of Obstetrics and Gynaecology, The Rosie Hospital and NIHR
Cambridge Comprehensive Biomedical Research Centre, University of Cambridge; Email: cema2@cam.ac.uk

Abstract
The theory of developmental programming suggests that diseases such as the metabolic syndrome may be ‘programmed’ by exposure
to adverse stimuli during early development. The developmental programming literature encompasses the study of a wide range of
suboptimal intrauterine environments in a variety of species and correlates these with diverse phenotypic outcomes in the offspring.
At a molecular level, a large number of variables have been measured and suggested as the basis of the programmed phenotype. The range
of both dependent and independent variables studied often makes the developmental programming literature complex to interpret
and the drawing of definitive conclusions difficult. A common, though under-explored, theme of many developmental programming
models is a sex difference in offspring outcomes. This holds true across a range of interventions, including dietary, hypoxic, and surgical
models. The molecular and phenotypic outcomes of adverse in utero conditions are often more prominent in male than female offspring,
although there is little consideration given to the basis for this observation in most studies. We review the evidence that maternal energy
investment in male and female conceptuses may not be equal and may be environment dependent. It is suggested that male and female
development could be viewed as separate processes from the time of conception, with differences in both timing and outcomes.
Reproduction (2013) 145 R1–R13

Introduction outcomes, for example, male sensitivity to nephrogenic


insult (Woods et al. 2005, Loria et al. 2007) and female
The theory of the Developmental Origins of Health and sensitivity to placental disruption (Gallou-Kabani et al.
Disease (DoHaD) suggests that diseases such as the 2010, Mao et al. 2010), these are not consistently
metabolic syndrome may be ‘programmed’ during observed between different models and between
development and early post-natal life. The theory different species.
suggests that insults to the conceptus during develop-
ment, for example, suboptimal maternal nutrition, may
manifest as type 2 diabetes, heart disease or hyper-
tension in later life (Hales & Barker 1992, Stanner et al. Sex differences in rodent models of developmental
1997, Ravelli et al. 1998). Many developmental programming
programming studies have found that male and female Sex differences in response to developmental program-
offspring exhibit different phenotypes following insults ming stimuli have been reported across a range of model
in utero (Table 1). This observed difference in outcomes organisms. Direct comparisons between male and
between male and female offspring has led many female offspring have been made predominantly in rats
researchers to target their efforts exclusively on offspring (Langley-Evans et al. 1996, Ozaki et al. 2001, Khan et al.
of one sex (Vieau et al. 2007). Despite wide recognition 2003, Nivoit et al. 2009, Reverte et al. 2011), but
of the importance of offspring sex in developmental sex-specific differences are also reported in mice
programming outcomes, the basis for the observed (Gallou-Kabani et al. 2010, Vickers et al. 2011; see
sexual dimorphism in ‘programming’ of the metabolic Table 1). There is evidence that male rat offspring are
syndrome is not understood. It is unclear what aspects of more susceptible to developmentally programmed
the differences between male and female development hypertension than female offspring in studies where the
give rise to the differential susceptibility to programming sexes are directly compared (Langley-Evans et al. 1996,
insults. Table 1 demonstrates that although there are Kwong et al. 2000, Dodic et al. 2002, Alexander 2003,
patterns suggesting differential susceptibilities to certain Woods et al. 2005, Maloney et al. 2011). This effect is
q 2013 Society for Reproduction and Fertility DOI: 10.1530/REP-11-0489
ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org
Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM
via free access
R2 C E Aiken and S E Ozanne

Table 1 Selected studies demonstrating a sex difference in developmentally programmed outcome in various species.
Reference Main result Species
Animal models
Male-specific outcomes
Langley-Evans et al. (1996) Low-protein diet gives elevated blood pressure in male offspring relative to females Rat
Kwong et al. (2000) Low-protein diet increases blood pressure in male offspring Rat
Ozaki et al. (2001) Nutrient restriction (30% reduction) increases blood pressure and vascular dysfunction Rat
more in male offspring
Dodic et al. (2002) Dexamethasone exposure gives increased blood pressure more in male offspring Sheep
Franco Mdo et al. (2002) Nutrient restriction (50%) reduces eNOS expression in aorta of male offspring Rat
Alexander (2003) Reduced uterine perfusion causes sustained increase in blood pressure in male offspring Rat
Ortiz et al. (2003) Antenatal dexamethasone injection persistently elevates blood pressure in male offspring Rat
Hemmings et al. (2005) Hypoxia gives increased myogenic tone in male offspring Rat
McMullen & Langley-Evans Low-protein diet gives elevated blood pressure, which is glucocorticoid dependent only Rat
(2005) in male offspring
Woods et al. (2005) Low-protein diet gives elevated blood pressure and reduced nephron number in Rat
male offspring
Zambrano et al. (2005) Isocaloric low-protein diet during lactation affects insulin sensitivity more in second- Rat
generation male offspring
Gilbert et al. (2007) Nutrient restriction impairs nephrogenesis in male offspring Sheep
Loria et al. (2007) Early post-natal ANGII blockade increases proteinuria and impairs urine concentrating Rat
ability in male offspring
Mueller & Bale (2007) Chronic variable stress exposure in early gestation decreases learning ability in Mouse
male offspring
Sinclair et al. (2007) Periconceptual reduction in methionine and B vitamins gives impaired blood pressure Sheep
in male offspring
Nivoit et al. (2009) High-fat and sugar diet increases insulin resistance in male offspring Rat
Tang et al. (2009) Prenatal betamethasone exposure decreased glomerular filtration rate and sodium Sheep
excretion in male offspring
Micke et al. (2010) Low-protein diet in the first trimester gives heavier male offspring in young adult life Cow
Reverte et al. (2011) Early post-natal ANGII blockade followed by high-protein diet leads to renal insufficiency Rat
in male offspring
Maloney et al. (2011) Methyl-deficient maternal diet periconceptually impairs glucose homoeostasis in Rat
male offspring
Female-specific outcomes
Lee & Rivier (1996) Ethanol exposure leads to exaggerated HPA stress response in young female offspring Rat
Khan et al. (2003) High-fat diet increases blood pressure in female offspring Rat
Wilcoxon et al. (2003) Ethanol exposure leads to decreased placental 11b-hydroxysteroid dehydrogenase-2 Rat
levels and left ventricular hypertrophy in female offspring
Manikkam et al. (2004) Prenatal testosterone exposure provokes post-natal catch-up growth in female Sheep
offspring only
Porter et al. (2007) High-salt diet preconceptually increases stress response in female offspring Rat
Padmavathi et al. (2009) Maternal zinc deficiency impairs glucose-induced insulin secretion in male but not Rat
female offspring
Mao et al. (2010) High-fat and low-fat diet produce up-regulation of placental genes more marked in Mouse
female offspring
Gallou-Kabani et al. (2010) High-fat diet leads to DNA hypomethylation in female placenta only Mouse
Rodriguez et al. (2011) Betamethasone exposure reduces associative learning more in female offspring Baboon
Vickers et al. (2011) Fructose exposure gives decreased placental weight, increased plasma leptin and Rat
glucose in female offspring
Epidemiology
Male-specific outcomes
Zaren et al. (2000) Maternal smoking decreases birth weight and head circumference proportionally more Human
in male offspring
Goldenberg et al. (2006) Placentas from preterm deliveries showed more lymphohistiocytic infiltration in Human
male offspring
Haley et al. (2006) Prenatal exposure to alcohol increases the heart rate of female infant offspring more than Human
males, but with the males showing a greater increase in plasma cortisol
Mingrone et al. (2008) Male offspring of obese mothers have higher levels of insulin secretion in response to GTT Human
Female-specific outcomes
Roseboom et al. (2001) Higher rates of obesity in female offspring conceived during famine Human
Clifton (2005) Maternal asthma in pregnancy causes altered placental glucocorticoid metabolism Human
and growth in female offspring
Stark et al. (2009) Antenatal betamethasone administration increased placental 11b-hydroxysteroid Human
dehydrogenase-2 activity in female offspring and this was positively correlated with
mean arterial blood pressure
Studies were included only if the results of both sexes were reported in the original paper. Studies were included if the effect of a developmental
programming insult was of greater statistical significance in one sex or was seen only in one sex. For animal models, only papers that specified an
intended developmental programming intervention were included.

Reproduction (2013) 145 R1–R13 www.reproduction-online.org

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
Sex differences in developmental programming R3

seen in rats with a range of interventions including placenta in both male and female offspring have reported
antenatal dexamethasone administration (Alexander that the placentas of female offspring are more readily
2003, Ortiz et al. 2003), prenatal low-protein diet affected by prenatal insults (Wilcoxon et al. 2003,
(Langley-Evans et al. 1996, Kwong et al. 2000, Woods Gallou-Kabani et al. 2010, Mao et al. 2010). It may be
et al. 2005) and uterine hypoperfusion (Alexander 2003). hypothesised that female offspring are more protected
The effect is not entirely consistent, however, as a from in utero insults by the relative ease with which
maternal high-fat diet has been reported to elevate blood the placenta adapts in the face of adverse conditions.
pressure in female offspring only (Khan et al. 2003). The The relative malleability of the female placenta is
attenuation of the endothelial response to acetylcholine also observed in other species, including the rat,
induced by this model was, however, present in both where ethanol exposure in utero leads to decreased
male and female offspring. In keeping with the 11b-hydroxysteroid dehydrogenase-2 activity in female
predominantly male hypertensive effects of prenatal offspring (Wilcoxon et al. 2003), and in human studies,
insults on development, studies have found more where placental up-regulation of 11b-hydroxysteroid
impaired renal function in male than in female rat dehydrogenase-2 activity in response to antenatal steroid
offspring (Woods et al. 2005, Gilbert et al. 2007, Loria administration occurs specifically in female offspring
et al. 2007, Reverte et al. 2011). Focusing on studies (Stark et al. 2009). 11b-Hydroxysteroid dehydrogenase-2
where the sexes are compared directly within the same is associated with improved physiological stability,
model, it has been suggested that an anatomical basis for particularly in infants born preterm, and it is therefore
impaired renal function can be seen in a direct reduction unsurprising to note that levels are decreased by a
of the number of nephrons in male offspring (Woods physiological insult such as alcohol consumption
et al. 2005). The greater susceptibility of male renal and increased by antenatal steroid administration
development to adverse stimuli is also demonstrated (which confers advantage in neonatal outcome; Stark
with post-natal intervention, when exposure to angio- et al. 2009).
tensin II receptor antagonists causes decreased nephron
number in both male and female rats but decreased
glomerular filtration rate and papillary volume only in Sex differences in larger mammals and humans
males (Saez et al. 2007). Sex-specific differences in offspring outcomes from
Impaired glucose tolerance and insulin resistance are developmental programming models are also demon-
also a common finding in rat offspring in developmental strated in farm animals, mainly sheep (Dodic et al. 2002,
programming studies. This feature has been noted in Manikkam et al. 2004, Gilbert et al. 2007, Sinclair et al.
both male and female offspring in various models 2007, Tang et al. 2009) and cows (Micke et al. 2010).
(Vickers et al. 2000, Petry et al. 2001, Simmons et al. A male offspring-predominant hypertensive effect,
2001, Seckl 2004, Ozanne et al. 2005). Where the sexes similar to that demonstrated in rat models, has been
are directly compared, the effect is observed in either the observed in sheep exposed to excess steroids in utero
male (Nivoit et al. 2009) or the female (Vickers et al. (Dodic et al. 2002) and in sheep fed a low-methionine
2011) offspring. It has further been suggested that and vitamin B diet in the periconceptual period (Sinclair
impaired insulin sensitivity is a male-specific finding in et al. 2007). In keeping with rodent data suggesting a
second-generation rat offspring when the dams of the F0 greater sensitivity of the developing renal system to
generation are exposed to isocaloric low-protein diet developmental insult in males than in females (Woods et
during lactation (Zambrano et al. 2005). Conflicting al. 2005, Saez et al. 2007), studies in sheep have also
results on offspring outcomes may be an effect related to demonstrated that prenatal steroid exposure reduces
the precise nature and timing of the intervention. glomerular filtration rate in male offspring (Tang et al.
Variations in the type of exposure, developmental age 2009) and nephrogenesis is impaired in male offspring
at which it was applied and age at which outcomes were exposed to nutrient restriction (Gilbert et al. 2007).
measured could all contribute to the differences in Growth patterns are addressed in larger mammals
outcomes observed. In the absence of further studies with the phenomenon of catch-up growth observed in
directly comparing offspring outcomes, systematic young adult male cows when exposed to low-protein
conclusions cannot be drawn. Despite the preponder- diet in the third trimester, with no significant difference
ance of developmental programming papers measuring in the carcass weights of females with the same exposure
outcomes related to hypertension, renal development (Micke et al. 2010). However, prenatal testosterone
and impaired glucose tolerance, other sex-specific exposure in sheep caused intrauterine growth retar-
differences in rodent studies specifically linked to dation (IUGR) in both sexes, but catch-up growth was
developmental programming are reported including reported in female offspring only (Manikkam et al. 2004).
learning ability (Mueller & Bale 2007), myogenic tone Human studies in developmental programming are
(Hemmings et al. 2005) and endothelial function (Franco by necessity limited to observational and epidemio-
Mdo et al. 2002). Studies of mouse models that have logical data; however, these do suggest that adverse
measured effects of developmental programming on the conditions during pregnancy have sexually dimorphic
www.reproduction-online.org Reproduction (2013) 145 R1–R13

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
R4 C E Aiken and S E Ozanne

effects. The developmental insults studied are wide- where this may reflect a nutritionally replete environ-
ranging, including maternal smoking (Zaren et al. 2000), ment or a maternal pathology that would compromise
antenatal steroid administration (Stark et al. 2009), the well-being of the pregnancy. It is clear that for
famine (Roseboom et al. 2001), asthma (Clifton 2005) propagation of the species, there must be survival of both
and obesity (Mingrone et al. 2008). These reflect the male and female offspring to reproductive years, but
results of animal models in showing effects on placental developmental programming could reflect subtle
function more in female offspring (Roseboom et al. 2001, differences in the long-term investment in each sex.
Clifton 2005, Stark et al. 2009), but growth restriction, Nonetheless, male conceptuses do survive in utero and
increased insulin secretion and plasma cortisol prefer- post-natally despite harsh conditions, and there is clearly
entially in males (Zaren et al. 2000, Haley et al. 2006, capacity to postulate that counter-mechanisms have
Mingrone et al. 2008). evolved to ensure survival of both sexes. On this basis,
In attempting to explain the difference in outcomes for we propose that females may be more sensitive and
male and female offspring, parallels may be drawn from adaptable to the intrauterine environment, whereas
fields other than developmental biology and molecular males may only be vulnerable and respond to the
biology, such as evolutionary biology. Evolutionary harshest of pre- and post-natal environments. The
biology traditionally views mammalian males as the evidence for the basis of a sex difference is examined
more ‘costly’ sex, and it may therefore not be energy- on both a species and an individual conceptus level, in
efficient for a mother to invest heavily in long-term an attempt to gain insight into the phenomenon.
protection of male conceptuses (Rickard et al. 2007,
Mathews et al. 2008). This type of effect may explain a
number of apparently anomalous results; for instance,
Evolutionary aspects of sexual dimorphism in
female infants of mildly asthmatic mothers demonstrate
a degree of IUGR, but this is ‘rescued’ after maternal developmental programming
exposure to inhaled steroids. The growth of male infants Mammalian sex differences may be considered on three
on the other hand is unaffected by mild asthma, but levels – primary reproductive characteristics; secondary
males have a high rate of in utero mortality and differences in males and females not directly linked to
severe IUGR if an acute exacerbation of asthma occurs reproduction (for example differences in muscle mass;
(Clifton 2005). Wells et al. 2010); and species-level differences in
Sexual dimorphism in terms of energy expenditure behaviours, hierarchies and mating strategies (Bouissou
and investment in conceptuses may be viewed as a 1983, Mank 2009). The first and second of these
species-level survival advantage to preserve precious differences are the areas with which developmental
resources at times of environmental stress in order to biology is primarily concerned; however, it may be
gain maximum chance of overall species survival. illuminating to additionally consider the third. Sexual
Adaptations in the offspring somatic tissues to environ- dimorphism is observed throughout the animal kingdom
mental stresses that are costly in terms of energy and is dependent on sex-limited gene expression
investment would therefore be made preferentially to (Williams & Carroll 2009). Patterns of sex-limited gene
female offspring, whose overall physiology and repro- expression and sex determination transcription factors
ductive tracts must be protected to ensure that they can are broadly similar in mammalian species but diverge
reproduce and withstand the stresses of pregnancy and across other classes and phyla (Williams & Carroll 2009).
lactation, whereas the male has only to protect his From an evolutionary biology point of view, the energy
germ cells and be fit to mate. This kind of adaptive invested by parents in male vs female offspring is thought
mechanism could be manifested in the changes to be influenced by the condition of the parents and
described in rat models in Table 1, i.e. lack of protection the post-natal environment faced by the offspring (Trivers
for male offspring in terms of renal tract development, & Willard 1973, Koskela et al. 2009).
and susceptibility to hypertensive disease (Langley-Evans The Trivers–Willard hypothesis (Trivers & Willard
et al. 1996, Kwong et al. 2000, Ozaki et al. 2001, 1973) states that in a species where there is a sex-
Alexander 2003, Woods et al. 2004, Loria et al. 2007), based difference in reproductive success, mothers with
where the somatic development of the male offspring is plentiful resources will be able to invest in the sex
‘killed’ to save maternal resources. Female offspring on with a reproductive disadvantage, whereas mothers
the other hand seem to have more placental adaptations facing an adverse environment will preferentially
to adverse conditions (Wilcoxon et al. 2003, Vickers produce offspring of the sex with a greater chance of
et al. 2011), which might reflect attempts to buffer reproductive success. If developmental programming is
and adapt to stressors, in order to preserve female considered as the molecular mechanism by which this
conceptuses in optimal condition and maximise the differential investment in offspring sex can be realised,
chances of successful pregnancy and raising of a litter. then the sexually dimorphic results of the various
The adaptive response to some interventions may not animal models of developmental programming may be
be clear-cut, for example to maternal hyperglycaemia, better understood.
Reproduction (2013) 145 R1–R13 www.reproduction-online.org

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
Sex differences in developmental programming R5

The application of the underlying principles of the environmental pressures. The weight of evidence for
Trivers–Willard hypothesis to the developmental pro- such an effect in the developmental programming
gramming hypothesis has several attractive elements. literature implies that a similar import should be
In particular, the inherent assumptions in both models attached to considering offspring sex when reviewing
are remarkably similar: first that maternal ‘condition’ and designing developmental programming studies.
relates to fetal ‘condition’, secondly that the effects Evidence for the Trivers–Willard effect is controversial
‘programmed’ by the in utero environment persist into in epidemiological studies of human populations, but
adult life and thirdly that the effects of advantageous several studies have found evidence of skewed sex ratios
conditions should be greater in the more reproductively linked to environmental conditions (Andersson &
variable sex (Trivers & Willard 1973). Detecting a sex Bergstrom 1998, Kanazawa 2007, Cameron & Dalerum
ratio skew in laboratory models of developmental 2009). The situation for monotocous as opposed to
programming is not straightforward, as many models polytocous mammals may differ substantially. If a
cull litters to a standardised number of males and Trivers–Willard effect does exist for monotocous mam-
females, or a set low number of single sex offspring in mals, including humans, it may also be more difficult to
order to maximise the plane of nutrition (Khan et al. detect in population studies, as there is no scope for a
2003, Loria et al. 2007, Chen et al. 2008, Tarry-Adkins subtle shift in sex ratio within a single pregnancy.
et al. 2010) but do not report pre-culling litter size or
gender distributions. Interestingly, however, a few
studies have shown that the sex ratio in rodent models Mechanistic aspects of sexual dimorphism in
can be altered by changing maternal diet (Rosenfeld developmental programming
et al. 2003, Rosenfeld & Roberts 2004), and a similar While it is useful to consider developmental program-
effect has been noted with calorie restriction (Meikle & ming at a species level, and to gain insight from an
Drickamer 1986) and increased fats in the maternal diet evolutionary perspective into the reasons behind the
(Fountain et al. 2008). It has been demonstrated that a utility of a developmental programming effect, it is also
mouse model utilising a maternal high-fat diet, which necessary to consider how sex-specific differences in
may be taken as a proxy for a nutrient-rich plentiful development occur at an individual level.
environment, produced litters skewed towards excess Three major factors give rise to sex differences in
males (male fraction 0.67) whereas a low-fat diet in the development: differences in patterns of development
same animals skewed the ratio towards excess female (genetic, transcriptional and morphological), differences
offspring (male fraction 0.39) (Rosenfeld et al. 2003). in timing of development and the influence of steroid
Calorie restriction in a mouse model gave results, hormone exposure during life in utero and post-natally.
consistent with the idea of a sex ratio skewed towards These factors combine to produce mature adult organ-
female offspring by acute dietary stress (Meikle & isms that are sexually dimorphic in terms of anatomy,
Drickamer 1986), but the sex ratio is not significantly physiology, reproductive capacity and behaviour (Fig. 1).
different after a long period of calorie restriction. This
adds weight to the idea of an adaptive process in
response to environmental stress. Genetic and morphological differences
The idea of maternal enhancement of the fitness of in development
one sex over the other depending on the post-natal
environment that the offspring will face is consistent with Evidence from rodent models
the differences observed in developmental programming From the earliest stages of preimplantation development,
models. Some of the sex differences described in the the sex of the embryo influences both growth patterns
developmental programming literature may be more and survival. In the mouse, cultured individual blas-
easily explained if viewed with respect to the evolution- tomeres from the two-cell embryo yield a higher rate of
ary basis rather than focusing on the molecular successful male than female conceptuses (Sato et al.
mechanism. If the Trivers–Willard hypothesis is 1995). Cultured preimplantation murine male embryos
re-written in terms more usually accessible to develop- show a faster rate of growth in vitro than do female
mental biologists, it postulates that the external environ- embryos (Valdivia et al. 1993). Fetal sex is determined by
ment is signalled to the conceptus by the mother via the presence or absence of a Y chromosome bearing the
the uterine environment. This signalling influences the Sry gene. In the mouse, this gene is responsible for the
phenotype of offspring produced to maximise chances onset of sexual dimorphism between embryonic (E) days
of species survival. This theory equates to the DoHaD 10.5–12.5. Sexual differentiation in the male initially
hypothesis and is a useful framework for considering the involves the interaction of SRY with SF1 to stimulate (via
apparent complexity of the observed sex differences in SOX9) Sertoli cell development in the genital ridges,
developmental programming phenotypes. More particu- which subsequently differentiate into testes (Sekido &
larly, the Trivers–Willard hypothesis specifies sexual Lovell-Badge 2008). However, there are other important
dimorphism as a key feature of the changes produced by central and peripheral regions expressing SRY, which
www.reproduction-online.org Reproduction (2013) 145 R1–R13

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
R6 C E Aiken and S E Ozanne

Zygote

Preimplantation embryo

Culture more successfully

Higher levels of antivirals


expressed

Transcriptional differences

Post-implantation

Sexually dimorphic adults Early post-natal life More susceptible to insults in culture
media, e.g. Ethanol

Increased rate of growth and higher


birth weight
Figure 1 Sex differences arising in male and female
Male sex is a significant risk factor
development from conception to the sexually
for adverse neonatal outcome dimorphic adult.

contribute to the development of a phenotypically male before expression of any overt sex-specific morphology,
or female neonate, including heart, liver and kidneys, survival may already be influenced in a sex-dependent
but particularly the brain and adrenal glands (Kopsida manner by the interaction of genetic factors and
et al. 2009). Cells derived from day 10.5 mouse embryos environmental conditions. By contrast, however, a
and grown in culture show similar patterns and rates of study of in vitro development of murine embryos in
cell division irrespective of chromosomal sex but differ elevated glucose concentrations failed to demonstrate
in their sensitivity to applied insults (e.g. ethanol) with any sex-specific effects, although total and trophecto-
female lineages showing higher rates of cell death derm cell numbers were reduced in all conceptuses
(Penaloza et al. 2009). (Bermejo-Alvarez et al. 2012). There is insufficient data
It is possible that female feto-placental units are more available to draw conclusions on whether glucose
responsive to adverse uterine environments at an early concentrations may influence development in vitro in a
stage. For instance, maternal high-fat diet during sex-specific manner in other species, but given the
pregnancy appears to directly influence placental difference in the findings of bovine and murine studies, it
methylation patterns only in female murine offspring may be expected that this will prove to be a species-
(Gallou-Kabani et al. 2010). This finding is consistent specific effect.
with the increased sensitivity of gene expression profiles In humans, male sex is a significant risk factor for
in female placentas to a maternal high-fat diet (Mao et al. adverse motor and cognitive outcomes in premature
2010). The decreased incidence of adverse outcomes in infants and for survival in neonatal intensive care units
female fetuses compared with males could be the (Stevenson et al. 2000), with 20% reduced survival of
consequence of a greater environmental adaptability, male preterm infants relative to females (Vatten &
and hence protection, by placental physiology. Skjaerven 2004). Recent evidence regarding the differing
outcomes of male and female premature births suggests
that feto-placental immune function may be sexually
Evidence from large mammal and human studies
dimorphic and may play a key role in influencing
There is evidence that development of male and female survival. Premature (!32 weeks) male neonates are
bovine conceptuses may be differently influenced by more likely to have a placenta showing evidence of
early culture conditions. In bovine blastocysts, the chronic inflammation and decidual lymphoplasmacytic
expression of antivirals in culture medium is twice as cell infiltration (Ghidini & Salafia 2005, Goldenberg
high in female conceptuses as in males, due to increased et al. 2006), whereas female placentas appear better
interferon t levels (Larson et al. 2001). This difference protected from inflammatory insult. The other major
has been postulated to give survival advantage to female difference influencing early life survival, and highlighted
embryos (Walker et al. 2009). In bovine conceptuses in many studies, is the degree of peripheral vasodilata-
cultured in vitro, increasing glucose concentrations in tion of the microvasculature, which is considerably
the culture medium led to an increased proportion of lower in female neonates (Stark et al. 2008). This
male vs female blastocysts surviving in culture to the difference confers a considerable survival benefit as it
blastocyst stage (Gutierrez-Adan et al. 2001, Larson et al. enables better cardiovascular stability and may be
2001). Thus, in the bovine preimplantation embryo, linked to the observations on immune function, via
Reproduction (2013) 145 R1–R13 www.reproduction-online.org

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
Sex differences in developmental programming R7

levels of circulating cytokines. These adaptations in the a marked difference in developmental programming of
female fetus require increased energy expenditure on offspring phenotypes when measured before and after
the feto-placental unit, which may be proportionally puberty. For example, in prenatal exposure to maternal
more costly to the mother in keeping with the Trivers– ethanol consumption (Lee & Rivier 1996), immature rat
Willard hypothesis. males and females show a dimorphic hypothalamo-
pituitary axis (HPA) response to immune stimuli, but
post-puberty, both show exaggerated ACTH release to
Timing of development the same stimuli. Spurious results may be obtained using
Human males and females are known to undergo experimental protocols that take measurements at an
development at different rates, both in utero and post- absolute time point after birth in young adult animals
natally up until the post-pubertal stage (Pedersen 1980, and which consequently fail to take account for the later
Davis et al. 1993, de Onis et al. 2009). Growth velocity sexual maturation of males, thereby observing an effect
may be critical to the extent to which an individual is that reflects pubertal status rather than primarily a sex
affected by an insult at any particular time point. A fetus difference.
growing faster in utero can be considered to have a In considering the applicability of these results to
greater effective exposure to a given insult than one that a human population, a further level of complexity is
undergoes fewer cell cycles during the period of added by the suggestion that a prenatal insult may
exposure. This may be the basis for the observation itself influence the timing of puberty, for example
from human studies that male infants of diabetic mothers children born at very low birth weight, whether at
show an increased incidence of fetal macrosomia term or prematurely, show early puberty, up to 2 years in
(weight at birth O4000 g, leading to increased risk of advance of their normal birth weight peers (Wehkalampi
adverse events at delivery and neonatal outcomes; et al. 2011). Hence, not only may the measured
Di Renzo et al. 2007), despite being exposed to the phenotype be influenced by the timing of the
same level of hyperglycaemia for the same absolute measurement but also by the nature and extent of the
time period as their female counterparts. insult applied.
Sex differences in temporal development have been
postulated from the time of conception itself, with the Reproductive aging in developmental programming
proposition that human male conceptuses are more In comparing phenotypes during later life, very few
likely to result at the extremes of the fertile period (James
developmental programming studies address ambient
2001, 2008). However, the empirical evidence for this
sex steroid levels at the time of sampling, yet a very
proposition is contentious (Gray et al. 1998). The theory
different outcome may be observed depending on
has been advanced that conception at either end of the
whether females still have the cardioprotective effect of
fertile window results in less ‘fit’ embryos, purporting to
oestrogen. Female rodents demonstrate cycle irregula-
explain why a higher number of spontaneously aborted
rities at around 9–12 months of age (Sokol et al. 1999,
conceptuses are male and advances an idea of male
Spencer et al. 2008), eventually becoming acyclic and
‘fragility’ extending through life (Kraemer 2000). Sexual
entering ‘estropause’ at 12–18 months of age (Van
dimorphism is also a feature of early post-implantation
Kempen et al. 2011). The key differences between
growth in mammalian conceptuses (Pedersen 1980).
menopause and estropause may be understood in
Later post-morphogenesis in utero growth is also faster in
terms of circulating oestrogen levels, which fall dramati-
human males with an average higher birth weight and
cally in human menopause, but are maintained at a
also a greater spread of birth weights (Clifton 2010);
higher ambient level in rodents (Maffucci & Gore 2006).
however, levels of b human chorionic gonadotrophin,
Many developmental programming studies have
which are responsible for maternal recognition and
sampling time points at around estropause (9–12
response to the pregnancy, are greater with a female than
months; Remmers et al. 2008) and yet do not consider
a male fetus (Cowans et al. 2009). Ultrasonographic
whether the effects of sex steroids on female tissues are
measurements of human growth in utero shows that the
consistent at this age, or indeed whether the results
slower rate of growth of the female fetus becomes
would have relevance to a human population.
significant at 28 weeks of gestation and increases
At the later extreme of life, aging and longevity have
towards term (Parker et al. 1984).
been studied in the developmental programming
literature, particularly with respect to telomere length
Timing of sexual development
(Jennings et al. 1999, Tarry-Adkins et al. 2008). It is
Sexual maturity is reached earlier in females than in known that telomere length in both the renal cortex and
males, an effect that is preserved across a range of the medulla of male rats is shorter than in females and
species from mice to humans, and may be correlated that telomere shortening is associated with increased
with expression of kisspeptin in the hypothalamus markers of cellular senescence (Tarry-Adkins et al.
(Clarkson & Herbison 2006, Kauffman 2010). There is 2006). In male rats, it has also been shown that prenatal
www.reproduction-online.org Reproduction (2013) 145 R1–R13

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
R8 C E Aiken and S E Ozanne

exposure to a maternal low-protein diet in rats shortens that oophorectomy in rats can narrow this gap in
telomeres prematurely in a number of organs incidence at an earlier age (Ojeda et al. 2007, 2008).
(Tarry-Adkins et al. 2008), but the effect is less well From a developmental point of view, male fetuses are
studied in females. exposed to higher levels of androgens in utero compared
In humans, differences in the lifespan of males and with female fetuses (Barry et al. 2010). In the human
females are well recognised, despite the lack of a fetus, it has been demonstrated that the testes can
demonstrated difference in telomere length at birth synthesise androgens from LDL-cholesterol by gesta-
(Okuda et al. 2002). Women have greater telomere tional week 10 (Carr et al. 1983). It is difficult to
lengths in later life than men in peripheral blood determine with accuracy fetal testosterone levels,
leukocytes (Benetos et al. 2001), and this difference particularly in the human where fetal and maternal
may be linked to oestrogen-dependent promotion of testosterone serum levels are poorly correlated (Scott
telomerase activity (Kyo et al. 1999). European women et al. 2009). The essential ‘masculinisation programming
undergo menopause at a median age of 54 years (Dratva window’ – when androgen levels become significant in
et al. 2009) and thereafter their general propensity to fetal serum – occurs shortly before the time at which
diseases of the metabolic syndrome changes consider- definitive male fetal phenotypic characteristics appear,
ably. A sex difference in developmental timing thus corresponding to E15.5–17.5 in the rat (Welsh et al.
extends throughout life and is worthy of consideration in 2008). If the molecular-level sex differences in rat
the design of developmental programming studies when developmental programming models are ascribed to
planning both exposure and measurement of phenotypic the difference in fetal testosterone exposure, it would be
outcomes. Spurious results may be obtained when reasonable to expect that sex differences should also
comparing males and females at the same absolute become detectable at around E15.5–17.5, or at least not
time from conception, but failing to take account of the detectable significantly before this time in the rat model.
different maturational stage that this represents. Particu- In female sheep offspring, it has been demonstrated that
larly in obtaining tissue samples from young adult in utero administration of testosterone to the mother
animals around the advent of sexual maturity, a sex produces pancreatic dysregulation and changes in liver
difference in pubertal stage is not often considered. metabolism (Hogg et al. 2011).
Similarly, in studies that involve senescent tissue, the A further consideration in polytocous organisms is the
normal rate of tissue aging in male and female tissues effect of differing levels of intrauterine sex steroid
and hence differences in for example telomere length exposure in the female offspring, dependent on intra-
and markers of oxidative damage should be considered. uterine position. Those females that developed in utero
between two male fetuses are known as ‘2M females’
and have higher testosterone exposure than ‘0M
Exposure to sex steroids females’, which develop between two female fetuses
Increased steroid exposure in utero has been demon- (vom Saal & Bronson 1980). Testosterone exposure of
strated to affect fetal and placental metabolism, nutrient 2M females has been shown to affect their subsequent
transfer and endocrine function (Milley 1996, Timmer- reproductive function (Vom Saal & Moyer 1985) and
man et al. 2000, Challis et al. 2002). Fetal glucocorticoid sexually dimorphic preoptic area development (Pei et al.
exposure is a strong candidate for involvement in 2006). The consideration of intrauterine position applies
programming of the metabolic syndrome and has been much less frequently to larger mammal and human
shown to affect development in a range of organisms populations that are not litter-bearing, but the differences
(Seckl 2008). It is unclear whether exposure to sex in physiology observed in 2M females adds weight to
steroids may have similar adverse effects. Evidence from the hypothesis that in utero exposure to sex steroids may
human populations suggests that the prevalence of profoundly influence developmental programming.
insulin insensitivity and polycystic ovarian syndrome is Much less work has gone into understanding the
higher in the presence of excess androgen hormones effects of female sex hormone exposure in utero, but it
(Veiga-Lopez et al. 2008, Moulana et al. 2011), and it has been shown that sex differences in insulin sensitivity
has been suggested that exposure to excess androgen in may be influenced by the effect of oestrogen as a leptin
childhood influences the development of the metabolic sensitiser on the fetal brain (Clegg et al. 2003, 2006).
syndrome and polycystic ovarian syndrome (Idkowiak
et al. 2011). In adult humans, male subjects show Sex-specific differences in epigenetic regulation
an increased propensity to at least some aspects of the Sex-specific differences in epigenetic modulations are
metabolic syndrome compared with females, possibly associated with developmentally programmed pheno-
as a primary effect of ambient sex steroid levels types in animal models (fully reviewed in Dunn et al.
(Regitz-Zagrosek et al. 2006). Epidemiological evidence (2011)). These effects are proposed to extend to second
shows that the sex difference in risk for cardiovascular generations of offspring in rats (Morgan & Bale 2011)
disease narrows considerably after the menopause and and can be induced by a variety of adverse stimuli,
Reproduction (2013) 145 R1–R13 www.reproduction-online.org

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
Sex differences in developmental programming R9

including maternal dietary and stress interventions pre- and post-natally, it may be argued that the sexes
(Dunn et al. 2011). should be treated by researchers as separate models.
In the female preimplantation embryo, there exists a In designing experiments, it is the goal of most
period during which transcription occurs from both X researchers to use the fewest animals and least resources
chromosomes, between the start of transcription from practicable to fully address their question. This leads to a
the embryonic genome and X inactivation. The absolute paucity of studies in which sexual dimorphism is built
extent of this window is species specific, lasting in the into the model, and yet this is a pertinent issue when
mouse between the eight-cell and blastocyst stages applying the results of developmental programming
(Gardner et al. 2010). The result of a period of expression studies to designing diagnostic, preventative and thera-
from both X chromosomes is a proteome considerably peutic measures.
different from that of the male (Epstein et al. 1978).
Differential expression of over 600 X-linked genes has
been shown in the mouse (Kobayashi et al. 2006), Declaration of interest
including dose-dependent up-regulation of hypoxan- The authors declare that there is no conflict of interest that
thine–guanine phosphoribosyltransferase (Kratzer & could be perceived as prejudicing the impartiality of the review
Gartler 1978) and glucose 6-phosphate dehydrogenase reported.
(Williams 1986). Persistence of these proteomic
changes after X inactivation could account for ongoing
differences in male and female growth patterns, as Funding
well as potentially determining their responsiveness to S E Ozanne is a British Heart Foundation Senior Fellow.
developmental insults.
Applying these considerations to larger mammals and
humans is more challenging – the timing and mechanism
of X inactivation is not well conserved across mamma- References
lian species (Okamoto & Heard 2009). The precise Alexander BT 2003 Placental insufficiency leads to development of
timing of X inactivation in human embryos is yet to be hypertension in growth-restricted offspring. Hypertension 41 457–462.
(doi:10.1161/01.HYP.0000053448.95913.3D)
fully determined (van den Berg et al. 2009). It remains Andersson R & Bergstrom S 1998 Is maternal malnutrition associated with a
uncertain how the contribution of X inactivation to low sex ratio at birth? Human Biology 70 1101–1106.
developmental programmed offspring phenotypes Barry JA, Kay AR, Navaratnarajah R, Iqbal S, Bamfo JE, David AL, Hines M
should be interpreted, particularly in view of the fact & Hardiman PJ 2010 Umbilical vein testosterone in female infants born
to mothers with polycystic ovary syndrome is elevated to male levels.
that not all genes on the X chromosome are silenced at Journal of Obstetrics and Gynaecology 30 444–446. (doi:10.3109/
X inactivation (Brown & Greally 2003). In humans, up 01443615.2010.485254)
to 15% of genes carried on the X chromosome appear Benetos A, Okuda K, Lajemi M, Kimura M, Thomas F, Skurnick J, Labat C,
Bean K & Aviv A 2001 Telomere length as an indicator of biological
to escape inactivation and hence a gene dosage effect aging: the gender effect and relation with pulse pressure and pulse wave
remains a plausible explanation for sex differences in velocity. Hypertension 37 381–385. (doi:10.1161/01.HYP.37.2.381)
developmental programming even after X inactivation van den Berg IM, Laven JS, Stevens M, Jonkers I, Galjaard RJ, Gribnau J &
has occurred (Brown & Greally 2003). van Doorninck JH 2009 X chromosome inactivation is initiated in human
preimplantation embryos. American Journal of Human Genetics 84
771–779. (doi:10.1016/j.ajhg.2009.05.003)
Bermejo-Alvarez P, Roberts RM & Rosenfeld CS 2012 Effect of glucose
Conclusions concentration during in vitro culture of mouse embryos on develop-
ment to blastocyst, success of embryo transfer, and litter sex ratio.
In the foregoing discussion, it is assumed that the sex Molecular Reproduction and Development 79 329–336. (doi:10.1002/
differences in developmental programming models are mrd.22028)
the result of the differential ability of the male and female Bouissou MF 1983 Androgens, aggressive behaviour and social relation-
fetus to respond to a particular stress; however, a further ships in higher mammals. Hormone Research 18 43–61. (doi:10.1159/
000179778)
question, which is difficult to address experimentally, Brown CJ & Greally JM 2003 A stain upon the silence: genes escaping X
is whether the mother’s response to a given stress is inactivation. Trends in Genetics 19 432–438. (doi:10.1016/S0168-
influenced by the sex of fetus she is carrying. Develop- 9525(03)00177-X)
Cameron EZ & Dalerum F 2009 A Trivers–Willard effect in contemporary
mental programming stresses are not normally applied to humans: male-biased sex ratios among billionaires. PLoS ONE 4 e4195.
the fetus directly, but particularly in the case of dietary (doi:10.1371/journal.pone.0004195)
interventions are mediated by the maternal physiology. Carr BR, Parker CR Jr, Ohashi M, MacDonald PC & Simpson ER 1983
It could therefore be postulated that all fetuses respond Regulation of human fetal testicular secretion of testosterone: low-
density lipoprotein-cholesterol and cholesterol synthesized de novo as
identically to the same insult, but the insult is buffered steroid precursor. American Journal of Obstetrics and Gynecology 146
by the mother in a way that depends on the sex of the 241–247.
fetus. This is a major point requiring clarification via Challis JR, Sloboda DM, Alfaidy N, Lye SJ, Gibb W, Patel FA, Whittle WL &
Newnham JP 2002 Prostaglandins and mechanisms of preterm birth.
further experimental work. Reproduction 124 1–17. (doi:10.1530/rep.0.1240001)
In view of the major temporal, spatial and biochemical Chen JH, Jones RH, Tarry-Adkins J, Smith NH & Ozanne SE 2008 Adverse
differences between male and female development, both effects of reduced oxygen tension on the proliferative capacity of rat

www.reproduction-online.org Reproduction (2013) 145 R1–R13

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
R10 C E Aiken and S E Ozanne

kidney and insulin-secreting cell lines involve DNA damage and stress infection and placental histologic findings in preterm births of males
responses. Experimental Cell Research 314 3075–3080. (doi:10.1016/ and females less than 32 weeks. American Journal of Obstetrics and
j.yexcr.2008.07.017) Gynecology 195 1533–1537. (doi:10.1016/j.ajog.2006.05.023)
Clarkson J & Herbison AE 2006 Postnatal development of kisspeptin Gray RH, Simpson JL, Bitto AC, Queenan JT, Li C, Kambic RT, Perez A,
neurons in mouse hypothalamus; sexual dimorphism and projections Mena P, Barbato M, Stevenson W et al. 1998 Sex ratio associated with
to gonadotropin-releasing hormone neurons. Endocrinology 147 timing of insemination and length of the follicular phase in planned and
5817–5825. (doi:10.1210/en.2006-0787) unplanned pregnancies during use of natural family planning. Human
Clegg DJ, Riedy CA, Smith KA, Benoit SC & Woods SC 2003 Differential Reproduction 13 1397–1400. (doi:10.1093/humrep/13.5.1397)
sensitivity to central leptin and insulin in male and female rats. Diabetes Gutierrez-Adan A, Granados J, Pintado B & De La Fuente J 2001 Influence
52 682–687. (doi:10.2337/diabetes.52.3.682) of glucose on the sex ratio of bovine IVM/IVF embryos cultured in vitro.
Clegg DJ, Brown LM, Woods SC & Benoit SC 2006 Gonadal hormones Reproduction, Fertility, and Development 13 361–365. (doi:10.1071/
determine sensitivity to central leptin and insulin. Diabetes 55 978–987. RD00039)
(doi:10.2337/diabetes.55.04.06.db05-1339) Hales CN & Barker DJ 1992 Type 2 (non-insulin-dependent) diabetes
Clifton VL 2005 Sexually dimorphic effects of maternal asthma during mellitus: the thrifty phenotype hypothesis. Diabetologia 35 595–601.
pregnancy on placental glucocorticoid metabolism and fetal growth. (doi:10.1007/BF00400248)
Cell and Tissue Research 322 63–71. (doi:10.1007/s00441-005-1117-5) Haley DW, Handmaker NS & Lowe J 2006 Infant stress reactivity and
Clifton VL 2010 Review: sex and the human placenta: mediating differential
prenatal alcohol exposure. Alcoholism, Clinical and Experimental
strategies of fetal growth and survival. Placenta 31 (Supplement 1)
Research 30 2055–2064. (doi:10.1111/j.1530-0277.2006.00251.x)
S33–S39. (doi:10.1016/j.placenta.2009.11.010)
Hemmings DG, Williams SJ & Davidge ST 2005 Increased myogenic tone
Cowans NJ, Stamatopoulou A, Maiz N, Spencer K & Nicolaides KH 2009
in 7-month-old adult male but not female offspring from rat dams
The impact of fetal gender on first trimester nuchal translucency and
exposed to hypoxia during pregnancy. American Journal of Physiology.
maternal serum free b-hCG and PAPP-A MoM in normal and trisomy 21
Heart and Circulatory Physiology 289 H674–H682. (doi:10.1152/
pregnancies. Prenatal Diagnosis 29 578–581. (doi:10.1002/pd.2246)
ajpheart.00191.2005)
Davis RO, Cutter GR, Goldenberg RL, Hoffman HJ, Cliver SP &
Brumfield CG 1993 Fetal biparietal diameter, head circumference, Hogg K, Wood C, McNeilly AS & Duncan WC 2011 The in utero
abdominal circumference and femur length. A comparison by race and programming effect of increased maternal androgens and a direct fetal
sex. Journal of Reproductive Medicine 38 201–206. intervention on liver and metabolic function in adult sheep. PLoS ONE 6
Di Renzo GC, Rosati A, Sarti RD, Cruciani L & Cutuli AM 2007 Does fetal e24877. (doi:10.1371/journal.pone.0024877)
sex affect pregnancy outcome? Gender Medicine 4 19–30. (doi:10.1016/ Idkowiak J, Lavery GG, Dhir V, Barrett TG, Stewart PM, Krone N & Arlt W
S1550-8579(07)80004-0) 2011 Premature adrenarche: novel lessons from early onset androgen
Dodic M, Abouantoun T, O’Connor A, Wintour EM & Moritz KM 2002 excess. European Journal of Endocrinology 165 189–207. (doi:10.1530/
Programming effects of short prenatal exposure to dexamethasone in EJE-11-0223)
sheep. Hypertension 40 729–734. (doi:10.1161/01.HYP.0000036455. James WH 2001 The fragile male. Male zygotes are often formed at
62159.7E) suboptimal times in fertile cycle. BMJ 322 617. (doi:10.1136/bmj.322.
Dratva J, Gomez Real F, Schindler C, Ackermann-Liebrich U, Gerbase MW, 7286.617)
Probst-Hensch NM, Svanes C, Omenaas ER, Neukirch F, Wjst M et al. James WH 2008 The variations of human sex ratio at birth with time of
2009 Is age at menopause increasing across Europe? Results on age at conception within the cycle, coital rate around the time of conception,
menopause and determinants from two population-based studies duration of time taken to achieve conception, and duration of gestation: a
Menopause 16 385–394. (doi:10.1097/gme.0b013e31818aefef) synthesis. Journal of Theoretical Biology 255 199–204. (doi:10.1016/j.
Dunn GA, Morgan CP & Bale TL 2011 Sex-specificity in transgenerational jtbi.2008.07.016)
epigenetic programming. Hormones and Behavior 59 290–295. (doi:10. Jennings BJ, Ozanne SE, Dorling MW & Hales CN 1999 Early growth
1016/j.yhbeh.2010.05.004) determines longevity in male rats and may be related to telomere
Epstein CJ, Smith S, Travis B & Tucker G 1978 Both X chromosomes shortening in the kidney. FEBS Letters 448 4–8. (doi:10.1016/S0014-
function before visible X-chromosome inactivation in female mouse 5793(99)00336-1)
embryos. Nature 274 500–503. (doi:10.1038/274500a0) Kanazawa S 2007 Beautiful parents have more daughters: a further
Fountain ED, Mao J, Whyte JJ, Mueller KE, Ellersieck MR, Will MJ, implication of the generalized Trivers–Willard hypothesis (gTWH).
Roberts RM, Macdonald R & Rosenfeld CS 2008 Effects of diets enriched Journal of Theoretical Biology 244 133–140. (doi:10.1016/j.jtbi.2006.
in omega-3 and omega-6 polyunsaturated fatty acids on offspring 07.017)
sex-ratio and maternal behavior in mice. Biology of Reproduction 78 Kauffman AS 2010 Gonadal and nongonadal regulation of sex differences
211–217. (doi:10.1095/biolreprod.107.065003) in hypothalamic Kiss1 neurones. Journal of Neuroendocrinology 22
Franco Mdo C, Arruda RM, Dantas AP, Kawamoto EM, Fortes ZB, 682–691. (doi:10.1111/j.1365-2826.2010.02030.x)
Scavone C, Carvalho MH, Tostes RC & Nigro D 2002 Intrauterine Khan IY, Taylor PD, Dekou V, Seed PT, Lakasing L, Graham D,
undernutrition: expression and activity of the endothelial nitric oxide Dominiczak AF, Hanson MA & Poston L 2003 Gender-linked
synthase in male and female adult offspring. Cardiovascular Research 56 hypertension in offspring of lard-fed pregnant rats. Hypertension 41
145–153. (doi:10.1016/S0008-6363(02)00508-4) 168–175. (doi:10.1161/01.HYP.0000047511.97879.FC)
Gallou-Kabani C, Gabory A, Tost J, Karimi M, Mayeur S, Lesage J, Kobayashi S, Isotani A, Mise N, Yamamoto M, Fujihara Y, Kaseda K,
Boudadi E, Gross MS, Taurelle J, Vige A et al. 2010 Sex- and diet-specific
Nakanishi T, Ikawa M, Hamada H, Abe K et al. 2006 Comparison of gene
changes of imprinted gene expression and DNA methylation in mouse
expression in male and female mouse blastocysts revealed imprinting
placenta under a high-fat diet. PLoS ONE 5 e14398. (doi:10.1371/
of the X-linked gene, Rhox5/Pem, at preimplantation stages. Current
journal.pone.0014398)
Biology 16 166–172. (doi:10.1016/j.cub.2005.11.071)
Gardner DK, Larman MG & Thouas GA 2010 Sex-related physiology of the
Kopsida E, Stergiakouli E, Lynn PM, Wilkinson LS & Davies W 2009 The role
preimplantation embryo. Molecular Human Reproduction 16 539–547.
of the Y chromosome in brain function. Open Neuroendocrinology
(doi:10.1093/molehr/gaq042)
Journal 2 20–30. (doi:10.2174/1876528900902010020)
Ghidini A & Salafia CM 2005 Histologic placental lesions in women
with recurrent preterm delivery. Acta Obstetricia et Gynecologica Koskela E, Mappes T, Niskanen T & Rutkowska J 2009 Maternal investment
Scandinavica 84 547–550. (doi:10.1111/j.0001-6349.2005.00694.x) in relation to sex ratio and offspring number in a small mammal – a case
Gilbert JS, Ford SP, Lang AL, Pahl LR, Drumhiller MC, Babcock SA, for Trivers and Willard theory? Journal of Animal Ecology 78 1007–1014.
Nathanielsz PW & Nijland MJ 2007 Nutrient restriction impairs (doi:10.1111/j.1365-2656.2009.01574.x)
nephrogenesis in a gender-specific manner in the ovine fetus. Pediatric Kraemer S 2000 The fragile male. BMJ 321 1609–1612. (doi:10.1136/bmj.
Research 61 42–47. (doi:10.1203/01.pdr.0000250208.09874.91) 321.7276.1609)
Goldenberg RL, Andrews WW, Faye-Petersen OM, Goepfert AR, Cliver SP Kratzer PG & Gartler SM 1978 HGPRT activity changes in preimplantation
& Hauth JC 2006 The Alabama Preterm Birth Study: intrauterine mouse embryos. Nature 274 503–504. (doi:10.1038/274503a0)

Reproduction (2013) 145 R1–R13 www.reproduction-online.org

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
Sex differences in developmental programming R11

Kwong WY, Wild AE, Roberts P, Willis AC & Fleming TP 2000 Maternal Mueller BR & Bale TL 2007 Early prenatal stress impact on coping strategies
undernutrition during the preimplantation period of rat development and learning performance is sex dependent. Physiology & Behavior 91
causes blastocyst abnormalities and programming of postnatal hyperten- 55–65. (doi:10.1016/j.physbeh.2007.01.017)
sion. Development 127 4195–4202. Nivoit P, Morens C, Van Assche FA, Jansen E, Poston L, Remacle C &
Kyo S, Takakura M, Kanaya T, Zhuo W, Fujimoto K, Nishio Y, Orimo A & Reusens B 2009 Established diet-induced obesity in female rats leads to
Inoue M 1999 Estrogen activates telomerase. Cancer Research 59 offspring hyperphagia, adiposity and insulin resistance. Diabetologia 52
5917–5921. 1133–1142. (doi:10.1007/s00125-009-1316-9)
Langley-Evans SC, Gardner DS & Jackson AA 1996 Maternal protein Ojeda NB, Grigore D, Yanes LL, Iliescu R, Robertson EB, Zhang H &
restriction influences the programming of the rat hypothalamic– Alexander BT 2007 Testosterone contributes to marked elevations in
pituitary–adrenal axis. Journal of Nutrition 126 1578–1585. mean arterial pressure in adult male intrauterine growth restricted
Larson MA, Kimura K, Kubisch HM & Roberts RM 2001 Sexual dimorphism offspring. American Journal of Physiology. Regulatory, Integrative
among bovine embryos in their ability to make the transition to expanded and Comparative Physiology 292 R758–R763. (doi:10.1152/ajpregu.
blastocyst and in the expression of the signaling molecule IFN-t. PNAS 00311.2006)
98 9677–9682. (doi:10.1073/pnas.171305398) Ojeda NB, Grigore D & Alexander BT 2008 Developmental programming
Lee S & Rivier C 1996 Gender differences in the effect of prenatal of hypertension: insight from animal models of nutritional mani-
alcohol exposure on the hypothalamic–pituitary–adrenal axis response pulation. Hypertension 52 44–50. (doi:10.1161/HYPERTENSIONAHA.
to immune signals. Psychoneuroendocrinology 21 145–155. (doi:10. 107.092890)
1016/0306-4530(95)00038-0) Okamoto I & Heard E 2009 Lessons from comparative analysis of
Loria A, Reverte V, Salazar F, Saez F, Llinas MT & Salazar FJ 2007 Sex and X-chromosome inactivation in mammals. Chromosome Research 17
age differences of renal function in rats with reduced ANG II activity 659–669. (doi:10.1007/s10577-009-9057-7)
during the nephrogenic period. American Journal of Physiology. Renal Okuda K, Bardeguez A, Gardner JP, Rodriguez P, Ganesh V, Kimura M,
Physiology 293 F506–F510. (doi:10.1152/ajprenal.00066.2007) Skurnick J, Awad G & Aviv A 2002 Telomere length in the newborn.
Maffucci JA & Gore AC 2006 Age-related changes in hormones and Pediatric Research 52 377–381. (doi:10.1203/00006450-200209000-
their receptors in animal models of female reproductive senescence. 00012)
In Handbook of Models for Human Aging, pp 533–552. Ed. MP Conn. de Onis M, Garza C, Onyango AW & Rolland-Cachera MF 2009 WHO
San Diego: Academic Press and Elsevier. growth standards for infants and young children. Archives de Pédiatrie
Maloney CA, Hay SM, Young LE, Sinclair KD & Rees WD 2011 A methyl- 16 47–53. (doi:10.1016/j.arcped.2008.10.010)
deficient diet fed to rat dams during the peri-conception period programs Ortiz LA, Quan A, Zarzar F, Weinberg A & Baum M 2003
glucose homeostasis in adult male but not female offspring. Journal of Prenatal dexamethasone programs hypertension and renal injury in
Nutrition 141 95–100. (doi:10.3945/jn.109.119453) the rat. Hypertension 41 328–334. (doi:10.1161/01.HYP.0000049763.
Manikkam M, Crespi EJ, Doop DD, Herkimer C, Lee JS, Yu S, Brown MB, 51269.51)
Foster DL & Padmanabhan V 2004 Fetal programming: prenatal Ozaki T, Nishina H, Hanson MA & Poston L 2001 Dietary restriction in
testosterone excess leads to fetal growth retardation and postnatal pregnant rats causes gender-related hypertension and vascular dysfunc-
catch-up growth in sheep. Endocrinology 145 790–798. (doi:10.1210/ tion in offspring. Journal of Physiology 530 141–152. (doi:10.1111/
en.2003-0478) j.1469-7793.2001.0141m.x)
Mank JE 2009 Sex chromosomes and the evolution of sexual dimorphism: Ozanne SE, Jensen CB, Tingey KJ, Storgaard H, Madsbad S & Vaag AA 2005
lessons from the genome. American Naturalist 173 141–150. (doi:10. Low birthweight is associated with specific changes in muscle insulin-
1086/595754) signalling protein expression. Diabetologia 48 547–552. (doi:10.1007/
Mao J, Zhang X, Sieli PT, Falduto MT, Torres KE & Rosenfeld CS 2010 s00125-005-1669-7)
Contrasting effects of different maternal diets on sexually dimorphic gene Padmavathi IJ, Kishore YD, Venu L, Ganeshan M, Harishankar N,
expression in the murine placenta. PNAS 107 5557–5562. (doi:10.1073/ Giridharan NV & Raghunath M 2009 Prenatal and perinatal zinc
pnas.1000440107) restriction: effects on body composition, glucose tolerance and insulin
Mathews F, Johnson PJ & Neil A 2008 You are what your mother eats: response in rat offspring. Experimental Physiology 94 761–769.
evidence for maternal preconception diet influencing foetal sex in (doi:10.1113/expphysiol.2008.045856)
humans. Proceedings. Biological Sciences/The Royal Society 275 Parker AJ, Davies P, Mayho AM & Newton JR 1984 The ultrasound
1661–1668. (doi:10.1098/rspb.2008.0105) estimation of sex-related variations of intrauterine growth. American
McMullen S & Langley-Evans SC 2005 Maternal low-protein diet in Journal of Obstetrics and Gynecology 149 665–669.
rat pregnancy programs blood pressure through sex-specific Pedersen JF 1980 Ultrasound evidence of sexual difference in fetal size in
mechanisms. American Journal of Physiology - Regulatory, Integrative first trimester. BMJ 281 1253. (doi:10.1136/bmj.281.6250.1253)
and Comparative Physiology 288 R85–R90. (doi:10.1152/ajpregu. Pei M, Matsuda K, Sakamoto H & Kawata M 2006 Intrauterine
00435.200400435.2004) proximity to male fetuses affects the morphology of the sexually
Meikle DB & Drickamer LC 1986 Food availability and secondary sex ratio dimorphic nucleus of the preoptic area in the adult rat brain. European
variation in wild and laboratory house mice (Mus musculus). Journal of Journal of Neuroscience 23 1234–1240. (doi:10.1111/j.1460-9568.
Reproduction and Fertility 78 587–591. (doi:10.1530/jrf.0.0780587) 2006.04661.x)
Micke GC, Sullivan TM, Gatford KL, Owens JA & Perry VE 2010 Nutrient Penaloza C, Estevez B, Orlanski S, Sikorska M, Walker R, Smith C, Smith B,
intake in the bovine during early and mid-gestation causes sex-specific Lockshin RA & Zakeri Z 2009 Sex of the cell dictates its response:
changes in progeny plasma IGF-I, liveweight, height and carcass traits. differential gene expression and sensitivity to cell death inducing stress in
Animal Reproduction Science 121 208–217. (doi:10.1016/j.anireprosci. male and female cells. FASEB Journal 23 1869–1879. (doi:10.1096/fj.08-
2010.05.017) 119388)
Milley JR 1996 Fetal substrate uptake during increased ovine fetal cortisol Petry CJ, Dorling MW, Pawlak DB, Ozanne SE & Hales CN 2001 Diabetes
concentration. American Journal of Physiology 271 E186–E191. in old male offspring of rat dams fed a reduced protein diet. International
Mingrone G, Manco M, Mora ME, Guidone C, Iaconelli A, Gniuli D, Journal of Experimental Diabetes Research 2 139–143. (doi:10.1155/
Leccesi L, Chiellini C & Ghirlanda G 2008 Influence of maternal obesity EDR.2001.139)
on insulin sensitivity and secretion in offspring. Diabetes Care 31 Porter JP, King SH & Honeycutt AD 2007 Prenatal high-salt diet in the
1872–1876. (doi:10.2337/dc08-0432) Sprague-Dawley rat programs blood pressure and heart rate hyper-
Morgan CP & Bale TL 2011 Early prenatal stress epigenetically programs responsiveness to stress in adult female offspring. American Journal of
dysmasculinization in second-generation offspring via the paternal Physiology - Regulatory, Integrative and Comparative Physiology 293
lineage. Journal of Neuroscience 31 11748–11755. (doi:10.1523/ R334–R342. (doi:10.1152/ajpregu.00887.2006)
JNEUROSCI.1887-11.2011) Ravelli AC, van der Meulen JH, Michels RP, Osmond C, Barker DJ,
Moulana M, Lima R & Reckelhoff JF 2011 Metabolic syndrome, androgens, Hales CN & Bleker OP 1998 Glucose tolerance in adults after prenatal
and hypertension. Current Hypertension Reports 13 158–162. (doi:10. exposure to famine. Lancet 351 173–177. (doi:10.1016/S0140-
1007/s11906-011-0184-0) 6736(97)07244-9)

www.reproduction-online.org Reproduction (2013) 145 R1–R13

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
R12 C E Aiken and S E Ozanne

Regitz-Zagrosek V, Lehmkuhl E & Weickert MO 2006 Gender differences Stanner SA, Bulmer K, Andres C, Lantseva OE, Borodina V, Poteen VV &
in the metabolic syndrome and their role for cardiovascular disease. Yudkin JS 1997 Does malnutrition in utero determine diabetes and
Clinical Research in Cardiology 95 136–147. (doi:10.1007/s00392-006- coronary heart disease in adulthood? Results from the Leningrad siege
0351-5) study, a cross sectional study BMJ 315 1342–1348. (doi:10.1136/bmj.
Remmers F, Verhagen LA, Adan RA & Delemarre-van de Waal HA 2008 315.7119.1342)
Hypothalamic neuropeptide expression of juvenile and middle-aged rats Stark MJ, Clifton VL & Wright IM 2008 Sex-specific differences
after early postnatal food restriction. Endocrinology 149 3617–3625. in peripheral microvascular blood flow in preterm infants. Pediatric
(doi:10.1210/en.2007-1388) Research 63 415–419. (doi:10.1203/01.pdr.0000304937.38669.63)
Reverte V, Tapia A, Moreno JM, Rodriguez L, Salazar F, Llinas MT & Stark MJ, Wright IM & Clifton VL 2009 Sex-specific alterations in placental
Salazar FJ 2011 Renal effects of prolonged high protein intake and COX2 11b-hydroxysteroid dehydrogenase 2 activity and early postnatal clinical
inhibition on hypertensive rats with altered renal development. course following antenatal betamethasone. American Journal of
American Journal of Physiology. Renal Physiology 301 F327–F333. Physiology. Regulatory, Integrative and Comparative Physiology 297
(doi:10.1152/ajprenal.00110.2011) R510–R514. (doi:10.1152/ajpregu.00175.2009)
Rickard IJ, Russell AF & Lummaa V 2007 Producing sons reduces lifetime Stevenson DK, Verter J, Fanaroff AA, Oh W, Ehrenkranz RA, Shankaran S,
reproductive success of subsequent offspring in pre-industrial Finns. Donovan EF, Wright LL, Lemons JA, Tyson JE et al. 2000 Sex differences
Proceedings. Biological Sciences/The Royal Society 274 2981–2988. in outcomes of very low birthweight infants: the newborn male
(doi:10.1098/rspb.2007.1051) disadvantage. Archives of Disease in Childhood. Fetal and Neonatal
Rodriguez JS, Zurcher NR, Keenan KE, Bartlett TQ, Nathanielsz PW & Edition 83 F182–F185. (doi:10.1136/fn.83.3.F182)
Nijland MJ 2011 Prenatal betamethasone exposure has sex specific Tang L, Carey LC, Bi J, Valego N, Sun X, Deibel P, Perrott J, Figueroa JP,
effects in reversal learning and attention in juvenile baboons. American Chappell MC & Rose JC 2009 Gender differences in the effects of
Journal of Obstetrics and Gynecology 204 e541–e545. (doi:10.1016/ antenatal betamethasone exposure on renal function in adult sheep.
j.ajog.2011.01.063) American Journal of Physiology. Regulatory, Integrative and Comparative
Roseboom TJ, van der Meulen JH, Ravelli AC, Osmond C, Barker DJ & Physiology 296 R309–R317. (doi:10.1152/ajpregu.90645.2008)
Bleker OP 2001 Effects of prenatal exposure to the Dutch famine on adult Tarry-Adkins JL, Ozanne SE, Norden A, Cherif H & Hales CN 2006 Lower
disease in later life: an overview. Molecular and Cellular Endocrinology antioxidant capacity and elevated p53 and p21 may be a link between
185 93–98. (doi:10.1016/S0303-7207(01)00721-3) gender disparity in renal telomere shortening, albuminuria, and long-
Rosenfeld CS & Roberts RM 2004 Maternal diet and other factors affecting evity. American Journal of Physiology. Renal Physiology 290 F509–F516.
offspring sex ratio: a review. Biology of Reproduction 71 1063–1070. (doi:10.1152/ajprenal.00215.2005)
(doi:10.1095/biolreprod.104.030890) Tarry-Adkins JL, Martin-Gronert MS, Chen JH, Cripps RL & Ozanne SE
Rosenfeld CS, Grimm KM, Livingston KA, Brokman AM, Lamberson WE & 2008 Maternal diet influences DNA damage, aortic telomere length,
Roberts RM 2003 Striking variation in the sex ratio of pups born to mice oxidative stress, and antioxidant defense capacity in rats. FASEB Journal
according to whether maternal diet is high in fat or carbohydrate. PNAS 22 2037–2044. (doi:10.1096/fj.07-099523)
100 4628–4632. (doi:10.1073/pnas.0330808100) Tarry-Adkins JL, Chen JH, Jones RH, Smith NH & Ozanne SE 2010 Poor
vom Saal FS & Bronson FH 1980 Sexual characteristics of adult female mice maternal nutrition leads to alterations in oxidative stress, antioxidant
are correlated with their blood testosterone levels during prenatal defense capacity, and markers of fibrosis in rat islets: potential underlying
development. Science 208 597–599. (doi:10.1126/science.7367881) mechanisms for development of the diabetic phenotype in later life.
Saez F, Castells MT, Zuasti A, Salazar F, Reverte V, Loria A & Salazar FJ FASEB Journal 24 2762–2771. (doi:10.1096/fj.10-156075)
2007 Sex differences in the renal changes elicited by angiotensin II Timmerman M, Teng C, Wilkening RB, Fennessey P, Battaglia FC &
blockade during the nephrogenic period. Hypertension 49 1429–1435. Meschia G 2000 Effect of dexamethasone on fetal hepatic glutamine–
(doi:10.1161/HYPERTENSIONAHA.107.087957) glutamate exchange. American Journal of Physiology. Endocrinology and
Sato E, Xian M, Valdivia RP & Toyoda Y 1995 Sex-linked differences in Metabolism 278 E839–E845.
developmental potential of single blastomeres from in vitro-fertilized Trivers RL & Willard DE 1973 Natural selection of parental ability to vary
2-cell stage mouse embryos. Hormone Research 44 (Suppl 2) 4–8. the sex ratio of offspring. Science 179 90–92. (doi:10.1126/science.179.
(doi:10.1159/000184653) 4068.90)
Scott HM, Mason JI & Sharpe RM 2009 Steroidogenesis in the fetal testis Valdivia RP, Kunieda T, Azuma S & Toyoda Y 1993 PCR sexing and
and its susceptibility to disruption by exogenous compounds. Endocrine developmental rate differences in preimplantation mouse embryos
Reviews 30 883–925. (doi:10.1210/er.2009-0016) fertilized and cultured in vitro. Molecular Reproduction and
Seckl JR 2004 Prenatal glucocorticoids and long-term programming. Development 35 121–126. (doi:10.1002/mrd.1080350204)
European Journal of Endocrinology 151 (Suppl 3) U49–U62. (doi:10. Van Kempen TA, Milner TA & Waters EM 2011 Accelerated ovarian failure:
1530/eje.0.151U049) a novel, chemically induced animal model of menopause. Brain
Seckl JR 2008 Glucocorticoids, developmental ‘programming’ and the Research 1379 176–187. (doi:10.1016/j.brainres.2010.12.064)
risk of affective dysfunction. Progress in Brain Research 167 17–34. Vatten LJ & Skjaerven R 2004 Offspring sex and pregnancy outcome by
(doi:10.1016/S0079-6123(07)67002-2) length of gestation. Early Human Development 76 47–54. (doi:10.1016/
Sekido R & Lovell-Badge R 2008 Sex determination involves synergistic j.earlhumdev.2003.10.006)
action of SRYand SF1 on a specific Sox9 enhancer. Nature 453 930–934. Veiga-Lopez A, Ye W, Phillips DJ, Herkimer C, Knight PG &
(doi:10.1038/nature06944) Padmanabhan V 2008 Developmental programming: deficits in
Simmons RA, Templeton LJ & Gertz SJ 2001 Intrauterine growth retardation reproductive hormone dynamics and ovulatory outcomes in prenatal,
leads to the development of type 2 diabetes in the rat. Diabetes 50 testosterone-treated sheep. Biology of Reproduction 78 636–647.
2279–2286. (doi:10.2337/diabetes.50.10.2279) (doi:10.1095/biolreprod.107.065904)
Sinclair KD, Allegrucci C, Singh R, Gardner DS, Sebastian S, Bispham J, Vickers MH, Breier BH, Cutfield WS, Hofman PL & Gluckman PD 2000
Thurston A, Huntley JF, Rees WD, Maloney CA et al. 2007 DNA Fetal origins of hyperphagia, obesity, and hypertension and postnatal
methylation, insulin resistance, and blood pressure in offspring amplification by hypercaloric nutrition. American Journal of Physiology.
determined by maternal periconceptional B vitamin and methionine Endocrinology and Metabolism 279 E83–E87.
status. PNAS 104 19351–19356. (doi:10.1073/pnas.0707258104) Vickers MH, Clayton ZE, Yap C & Sloboda DM 2011 Maternal fructose
Sokol RZ, Okuda H, Stanczyk FZ, Wolfe GW, Delaney JC & Chapin RE intake during pregnancy and lactation alters placental growth and leads
1999 Normative reproductive indices for male and female adult to sex-specific changes in fetal and neonatal endocrine function.
Sprague-Dawley rats. Contraception 59 203–207. (doi:10.1016/S0010- Endocrinology 152 1378–1387. (doi:10.1210/en.2010-1093)
7824(99)00017-7) Vieau D, Sebaai N, Leonhardt M, Dutriez-Casteloot I, Molendi-Coste O,
Spencer JL, Waters EM, Milner TA & McEwen BS 2008 Estrous cycle Laborie C, Breton C, Deloof S & Lesage J 2007 HPA axis pro-
regulates activation of hippocampal Akt, LIM kinase, and neurotrophin gramming by maternal undernutrition in the male rat offspring.
receptors in C57BL/6 mice. Neuroscience 155 1106–1119. (doi:10. Psychoneuroendocrinology 32 (Suppl 1) S16–S20. (doi:10.1016/j.psy
1016/j.neuroscience.2008.05.049) neuen.2007.03.014)

Reproduction (2013) 145 R1–R13 www.reproduction-online.org

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access
Sex differences in developmental programming R13

Vom Saal FS & Moyer CL 1985 Prenatal effects on reproductive capacity Williams TM & Carroll SB 2009 Genetic and molecular insights into
during aging in female mice. Biology of Reproduction 32 1116–1126. the development and evolution of sexual dimorphism. Nature Reviews.
(doi:10.1095/biolreprod32.5.1116) Genetics 10 797–804. (doi:10.1038/nrg2687)
Walker AM, Kimura K & Roberts RM 2009 Expression of bovine interferon-t Woods LL, Weeks DA & Rasch R 2004 Programming of adult
variants according to sex and age of conceptuses. Theriogenology 72 blood pressure by maternal protein restriction: role of nephrogenesis.
44–53. (doi:10.1016/j.theriogenology.2009.01.017) Kidney International 65 1339–1348. (doi:10.1111/j.1523-1755.2004.
Wehkalampi K, Hovi P, Dunkel L, Strang-Karlsson S, Jarvenpaa AL, 00511.x)
Eriksson JG, Andersson S & Kajantie E 2011 Advanced pubertal growth Woods LL, Ingelfinger JR & Rasch R 2005 Modest maternal
spurt in subjects born preterm: the Helsinki study of very low birth weight protein restriction fails to program adult hypertension in female rats.
adults. Journal of Clinical Endocrinology and Metabolism 96 525–533. American Journal of Physiology. Regulatory, Integrative and Comparative
(doi:10.1210/jc.2010-1523) Physiology 289 R1131–R1136. (doi:10.1152/ajpregu.00037.2003)
Wells JC, Williams JE, Chomtho S, Darch T, Grijalva-Eternod C, Kennedy K, Zambrano E, Martinez-Samayoa PM, Bautista CJ, Deas M, Guillen L,
Haroun D, Wilson C, Cole TJ & Fewtrell MS 2010 Pediatric reference Rodriguez-Gonzalez GL, Guzman C, Larrea F & Nathanielsz PW 2005
data for lean tissue properties: density and hydration from age 5 to 20 y. Sex differences in transgenerational alterations of growth and metab-
American Journal of Clinical Nutrition 91 610–618. (doi:10.3945/ajcn. olism in progeny (F2) of female offspring (F1) of rats fed a low protein diet
2009.28428) during pregnancy and lactation. Journal of Physiology 566 225–236.
Welsh M, Saunders PT, Fisken M, Scott HM, Hutchison GR, Smith LB & (doi:10.1113/jphysiol.2005.086462)
Sharpe RM 2008 Identification in rats of a programming window for Zaren B, Lindmark G & Bakketeig L 2000 Maternal smoking affects fetal
reproductive tract masculinization, disruption of which leads to growth more in the male fetus. Paediatric and Perinatal Epidemiology 14
hypospadias and cryptorchidism. Journal of Clinical Investigation 118 118–126. (doi:10.1046/j.1365-3016.2000.00247.x)
1479–1490. (doi:10.1172/JCI34241)
Wilcoxon JS, Schwartz J, Aird F & Redei EE 2003 Sexually dimorphic effects of
maternal alcohol intake and adrenalectomy on left ventricular hypertrophy
in rat offspring. American Journal of Physiology. Endocrinology and Received 21 December 2011
Metabolism 285 E31–E39. (doi:10.1152/ajpendo.00552.2002)
First decision 2 February 2012
Williams TJ 1986 A technique for sexing mouse embryos by a visual
colorimetric assay of the X-linked enzyme, glucose 6-phosphate dehydro- Revised manuscript received 1 October 2012
genase. Theriogenology 25 733–739. (doi:10.1016/0093-691X(86)90131-7) Accepted 15 October 2012

www.reproduction-online.org Reproduction (2013) 145 R1–R13

Downloaded from Bioscientifica.com at 11/25/2018 10:35:05PM


via free access

You might also like