Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

J Physiol 590.

21 (2012) pp 5503–5518 5503

Maternal high-fat diet induces obesity and adrenal and


thyroid dysfunction in male rat offspring at weaning
J. G. Franco1 , T. P. Fernandes2 , C. P. D. Rocha2 , C. Calviño2 , C. C. Pazos-Moura2 , P. C. Lisboa1 ,
E. G. Moura1 and I. H. Trevenzoli1,2
1
Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Brazil
2
Laboratory of Molecular Endocrinology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Brazil

Key points
• Perinatal maternal high-fat diet changes milk composition, resulting in obesity and hyper-
glycaemia in male offspring at weaning.
• Offspring obesity is associated with hyperleptinaemia and changes in the central leptin
The Journal of Physiology

signalling pathway in the hypothalamic arcuate nucleus.


• Maternal high-fat diet increased adrenal catecholamines in offspring but reduced liver and
adipose tissue adrenoreceptors, thereby contributing to increased adiposity in these animals.
• Early obesity and hyperleptinaemia in offspring may have a stimulatory effect on the
hypothalamus–pituitary–thyroid axis as an adaptive response to the positive energy balance.
• Both catecholamines and thyroid hormones may impact cardiovascular function, thereby
contributing to the development of hypertension.

Abstract Maternal nutritional status affects the future development of offspring. Both under-
nutrition and overnutrition in critical periods of life (gestation or lactation) may cause several
hormonal changes in the pups and programme obesity in the adult offspring. We have
shown that hyperleptinaemia during lactation results in central leptin resistance, higher adrenal
catecholamine secretion, hyperthyroidism, and higher blood pressure and heart rate in the adult
rats. Here, we evaluated the effect of a maternal isocaloric high-fat diet on breast milk composition
and its impact on leptinaemia, energy metabolism, and adrenal and thyroid function of the
offspring at weaning. We hypothesised that the altered source of fat in the maternal diet even
under normal calorie intake would disturb the metabolism of the offspring. Female Wistar
rats were fed a normal (9% fat; C group) or high-fat diet (29% fat as lard; HF group) for
8 weeks before mating and during pregnancy and lactation. HF mothers presented increased
total body fat content after 8 weeks (+27%, P < 0.05) and a similar fat content at the end of
lactation. In consequence, the breast milk from the HF group had higher concentration of protein
(+18%, P < 0.05), cholesterol (+52%, P < 0.05) and triglycerides (+86%, P < 0.05). At weaning,
HF offspring had increased body weight (+53%, P < 0.05) and adiposity (2 fold, P < 0.05),
which was associated with lower β3-adrenoreceptor content in adipose tissue (−40%, P < 0.05).
The offspring also presented hyperglycaemia (+30%, P < 0.05) and hyperleptinaemia (+62%,
P < 0.05). In the leptin signalling pathway in the hypothalamus, we found lower p-STAT3/STAT3
(−40%, P < 0.05) and SOCS3 (−55%, P < 0.05) content in the arcuate nucleus, suggesting leptin
resistance. HF offspring also had higher adrenal catecholamine content (+17%, P < 0.05), liver
glycogen content (+50%, P < 0.05) and hyperactivity of the thyroid axis at weaning. Our results
suggest that a high fat diet increases maternal body fat and this additional energy is transferred to
the offspring during lactation, since at weaning the dams had normal fat and the pups were obese.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society DOI: 10.1113/jphysiol.2012.240655
5504 J. G. Franco and others J Physiol 590.21

The higher fat and protein concentrations in the breast milk seemed to induce early overnutrition
in the HF offspring. In addition to storing energy as fat, the HF offspring had a larger reserve
of glycogen and hyperglycaemia that may have resulted from increased gluconeogenesis. Hyper-
leptinaemia may stimulate both adrenal medullary and thyroid function, which may contribute to
the development of cardiovascular diseases. These early changes induced by the maternal high-fat
diet may contribute to development of metabolic syndrome.
(Resubmitted 10 July 2012; accepted 3 August 2012; first published online 6 August 2012)
Corresponding author I. H. Trevenzoli: Laboratório de Endocrinologia Molecular, Instituto de Biofı́sica Carlos Chagas
Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, sl G0-16, Cidade Universitária–Rio de
Janeiro, RJ, 21941-902 Brazil. Email: haraisis@biof.ufrj.br

Abbreviations AR, adrenoreceptor; BW, body weight; CART, Cocaine - and amphetamine - regulated transcript;
DEXA, dual energy X-ray absorptiometry; FFA, free fatty acid; FT4, free T4; JAK2, Janus tyrosine kinase 2; OBRb, long
form of leptin receptor; PNMT, phenylethanolamine-N -methyl transferase; p-STAT3, phosphorylated signal transducer
and activator of transcription 3; PVN, paraventricular hypothalamic nucleus; SOCS3, suppressor of cytokine signalling
3; STAT3, signal transducer and activator of transcription 3; TH, tyrosine hydroxylase; TT3, total T3.

Introduction as a consequence of the altered secretion of several


hormones and factors, such as increased leptin and serum
Epidemiological and experimental evidence has shown free fatty acids (FFAs) and decreased adiponectin (Ahima
that malnutrition in early life is associated with the & Lazar, 2008).
development of chronic diseases at adulthood, such as Leptin is secreted mainly by white adipocytes and
type 2 diabetes and hypertension (Barker et al. 2002; binds to the long form of leptin receptor (OBRb) in
Moura & Passos, 2005; Moura et al. 2008). During the the hypothalamus, reducing appetite and stimulating
gestation and lactation periods, offspring are extremely energy expenditure through the Janus tyrosine kinase 2
sensitive to nutritional, hormonal and environmental (JAK2)–signal transducer and activator of transcription
changes that may result in altered physiology of several 3 (STAT3) pathway. There are also leptin receptors in
tissues and systems throughout life (Simmons, 2005). This other tissues, such as mammary glands, placenta, pituitary
phenomenon is named ‘programming’, and its origins are gland, adrenals, thyroid and components of the cardio-
related to epigenetic modifications – DNA methylation vascular system (Bjorbaek & Kahn, 2004), suggesting
and histone acetylation – during critical periods of life that this hormone may also regulate metabolism peri-
(Burdge et al. 2007). pherally. Serum leptin level is positively correlated with
Obesity is the most frequent characteristic programmed adiposity. However, obese humans and animals usually
in humans and experimental models (Spencer, 2012). Both do not respond to the anorexigenic effect of leptin
maternal undernutrition and overnutrition during the because of the development of leptin resistance in the
perinatal period may programme obesity development hypothalamus.
in the offspring in rodent models (Teixeira et al. We have shown that hyperleptinaemia during the
2002; Rodrigues et al. 2009). Rats whose mothers were first 10 days of lactation promotes obesity, central leptin
undernourished during gestation have low birth weight. resistance and higher levels of catecholamines, thyroid
However, they display catch-up growth and become hormones, and blood pressure at adulthood (Toste
obese at adulthood (Martin-Gronert & Ozanne, 2010). et al. 2006a,b; Trevenzoli et al. 2007). Currently, a
When dams are protein-restricted only during lactation, large number of women of reproductive age are over-
the offspring have the opposite phenotype (Fagundes weight or obese (Callaway et al. 2006; Kim et al. 2007).
et al. 2009). On the other hand, maternal obesity or Here, we hypothesised that maternal consumption of a
high-fat diet consumption during the perinatal period also high-fat diet during the perinatal period and increased
programmes obesity at adulthood (Samuelsson et al. 2008; maternal adiposity would affect leptinaemia and energy
Sullivan et al. 2011). These phenotypes may be associated metabolism in their offspring, increasing the risk for
with changes in leptin levels in early life that may disturb developing obesity and cardiovascular failure. Therefore,
the development of important regulatory sites of energy we aimed to elucidate the impact of maternal consumption
metabolism, such as the hypothalamus (Bouret & Simerly, of an isocaloric high-fat diet during critical periods
2006). of development, such as gestation and lactation, on
Obesity is characterised by an imbalance between breast milk composition and on the adiposity, glucose
energy intake and expenditure that results in the homeostasis, leptinaemia and central leptin sensitivity of
enlargement of adipose tissue mass. Adipocyte hyper- the offspring at weaning to determine if any of these
trophy is associated with many metabolic changes, mainly


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
J Physiol 590.21 Maternal high fat diet programmes adrenal and thyroid function 5505

At birth, all of the litters were weighed and adjusted to


Table 1. Composition of the control and high-fat diets
six males for each dam, a number that maximises lactation
Macronutrients kcal %kcal performance (Fishbeck & Rasmussen, 1987). Litters that
Control diet (amount per kg) did not have at least six male pups were discarded.
Protein 220 g 880 22.6 At weaning, the dams again underwent DEXA, which
Lipids 40 g 360 9.4 was followed by killing using a rodent guillotine. The
Carbohydrates 660 g 2,640 68 glycaemia of the pups was measured, and then they
Total 1000 g 3,880 100 were killed by cardiac puncture under anaesthesia (55 mg
High-fat diet (amount per kg) (kg BW)−1 ketamine and 100 mg (kg BW)−1 xylazine)
Protein 169.6 g 678.4 17.3 for sample collection. Blood samples were centrifuged
Lipids 214.9 g 1124 28.6 (1000 g, 4◦ C, 20 min), and serum was kept individually
Carbohydrates 530.3 g 2121 54.1 at −20◦ C until hormone quantification assays. The left
Total 1092 g 3923 100 adrenal glands were dissected and kept in 10% acetic
acid solution at −20◦ C for catecholamine quantification.
The right adrenal glands were kept at −80◦ C to measure
the catecholamine-synthesising enzymes tyrosine hydro-
xylase (TH) and phenylethanolamine-N -methyl trans-
parameters were associated with altered adrenal or thyroid ferase (PNMT). Liver samples were collected for glycogen
function. quantification and analysis of β2 -adrenoreceptor (β2AR)
content by Western blotting. We dissected and weighed
three representative depots of white adipose tissue
Methods (retroperitoneal, epididymal and inguinal) to analyse
adiposity in the pups. Samples from the inguinal region
Animals and diets
were taken to analyse leptin, β2AR, and β3AR expression.
The use of animals in our experimental design was The brain was collected for dissection of the arcuate
approved by the Animal Care and Use Committee of the nucleus of the hypothalamus, which was used to quantify
Carlos Chagas Filho Biophysics Institute (process IBCCF leptin signalling proteins. The hearts of the offspring were
114). The handling of the experimental animals followed also weighed.
the principles adopted in the UK and Brazil according For each experimental procedure, animals from at least
the Brazilian Law no. 11.794/2008 (Drummond, 2009; six different litters per group were used to avoid litter
Marques et al. 2009). effects.
Female Wistar rats were kept in an environmentally
controlled room (23 ± 2◦ C and a 12 h light–dark cycle
with lights on from 07.00 h to 19.00 h). Nulliparous
Biochemical and hormonal milk analysis
60-day-old female rats were divided into two groups: the
control group (C), which received a standard diet for Lactose, protein, cholesterol, triglycerides and leptin were
rodents (9% of the calories as fat), and a high-fat group measured in milk samples from the C and HF dams in the
(HF), which received a high-fat diet (28% of the calories as middle (day 11) and at the end (day 21) of lactation (Troina
fat). In the high-fat diet, lard was used as fat source (mainly et al. 2010). Lactating rats were separated from their pups
saturated fat) and we also added 1% soy oil to provide for 2 h and then injected subcutaneously with 5 IU ml−1 of
the minimal amount of n3 fatty acid (mainly unsaturated oxytocin (Eurofarm, São Paulo, SP, Brazil). After 20 min,
fat) for adequate development of rats. The macronutrient the dams were lightly anaesthetised (55 mg (kg BW)−1
composition of the control and high-fat diets is shown in ketamine and 100 mg (kg BW)−1 xylazine), and milk was
Table 1. Both diets contained approximately 3.9 kcal g−1 extracted by gently squeezing the thoracic and abdominal
and followed the AIN-93 recommendations for micro- teats. Lactose content was estimated by a colorimetric
nutrients (Reeves et al. 1993). The female rats were method using commercial lactose (Sigma, St Louis, MO,
fed these diets beginning 8 weeks before mating and USA) as a standard. Protein content was measured
throughout gestation and lactation. After the first 8 weeks by the colorimetric Peterson method using bovine
on the diets, the body composition of the dams was serum albumin (Sigma) as a standard. Total cholesterol
evaluated by dual energy X-ray absorptiometry (DEXA) and triglycerides were measured by an enzymatic and
to confirm the effect of the HF diet in increasing body fat colorimetric method using a commercial kit (Bioclin,
content. Next, the dams were mated in a 3 to 1 ratio. After Belo Horizonte, MG, Brazil). The leptin concentration
mating, pregnant rats were housed in individual standard of the milk was evaluated using a rat leptin-specific radio-
rat cages with free access to water and food until the pups’ immunoassay (RIA) kit with a sensitivity of 0.5 ng ml−1
birth. The dams were weighed weekly. (Linco Research, Inc., St Charles, MO, USA). For these


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
5506 J. G. Franco and others J Physiol 590.21

analyses, we used six mothers per group for each time measurements, we used nine C-offspring pups and 10
point. HF-offspring pups.

Microdissection of paraventricular (PVN) and arcuate


Hormones and free fatty acid (FFA) measurements
(Arc) hypothalamic nuclei
Total serum adiponectin was measured by ELISA
PVN and Arc tissues were obtained by the punch technique
(Millipore, Billerica, MA, USA) with an assay sensitivity of
from thick coronal brain sections cut in a cryostat using the
0.155 ng ml−1 . Serum leptin, insulin, corticosterone, total
bregma as reference (Palkovits, 1973; Paxinos & Watson,
T3 (TT3) and free T4 (FT4) were measured using specific
2007; Helena et al. 2009). Briefly, two subsequent sections
commercial RIA kits with the following assay sensitivities:
of 1000 μM were made: from −0.6 mm to −1.6 mm for
leptin: 0.5 ng ml−1 (Linco Research, Inc.); insulin:
PVN microdissection, and from −1.6 mm to −2.6 mm
0.1 ng ml−1 (ICN Pharmaceuticals Inc., Orangeburg, NY,
for microdissection of the Arc. The PVN was dissected
USA); corticosterone: 50 ng ml−1 (Zen-bio Inc., Research
from the first section in one punch, obtained with a
Triangle Park, NC, USA); and TT3 and FT4: 6.7 ng dl−1
1.5 mm-diameter round needle with the top centred
and 0.045 ng dl−1 , respectively (Coat-A-Coat, DPC, Los
in the third ventricle. The Arc was dissected from the
Angeles, CA, USA). Serum TSH was measured by specific
second section using a 2 mm ‘square puncher’ needle,
rat RIA employing reagents supplied by the National
centred on the third ventricle, approximately 1 mm dorsal
Hormone and Peptide Program (Torrance, CA, USA) as
to the base of the brain. Immediately afterwards, the
previously described (Ortiga-Carvalho et al. 2002). The
tissue samples were kept at −80◦ C until Western blotting
intra-assay coefficients of variance of the assays for all
assays.
hormones were in the range of 4–7%. Serum FFA was
measured using a colorimetric commercial kit (Zen-bio
Inc.). For each parameter that was evaluated, all of the
Western blotting
measurements were performed in a single assay. For
these assays, we used nine C-offspring pups and ten We used the western blotting assay to detect specific
HF-offspring pups. proteins in various tissues: Arc, PVN, adipose tissue, liver
and adrenal. Samples of each tissue were homogenised in
a specific way, following previous protocols with minor
modifications (Trevenzoli et al. 2010a).
Real-time PCR
To analyse the leptin signalling pathway and TRH in
Leptin gene expression was quantified as previously hypothalamic punch samples, we used two nuclei pools
described (Nobre et al. 2011). Total RNA was isolated to obtain each sample and achieve an adequate protein
from inguinal adipose tissue using a commercial kit concentration in the tissue extracts. The samples were
(RNeasy lipid tissue mini kit, Qiagen, Hilden Germany). homogenised in 100 μl of ice-cold lysis buffer (50 mM
For quantitative real-time RT-PCR analysis, reverse Hepes, 1 mM MgCl2 , 10 mM EDTA, 1% Triton X-100
transcription was carried out on 1 μg of total RNA for pH 6.4) containing protease inhibitors (Complete, Roche
all tissues using a SuperScript III kit. The products were Diagnostics), using an automatic ultrasonic processor
amplified in an Applied Biosystems 7500 real-time PCR (2 × 10 s pulse on and 20 s pulse off ). The total protein
System (Life Technologies Co., Frederick, MD, USA) using concentration of the homogenates was determined using
SYBR Green PCR Master Mix (Applied BioSystems, Foster the BCA Protein Assay Kit (Thermo Scientific, Rockford,
City, CA, USA). Briefly, after initial denaturation at 50◦ C IL, USA).
for 2 min and 95◦ C for 10 min, the reactions were cycled 40 To analyse the catecholamine-synthesising pathway,
times using the following parameters for the ob gene: 95◦ C each right adrenal was homogenised manually in 500 μl
for 15 s, 53◦ C for 30 s and 70◦ C for 45 s. Product purity of lysis buffer. The homogenates were centrifuged (1120 g,
was confirmed by agarose gel analysis. Relative mRNA 4◦ C, 5 min), and the supernatants were collected for total
levels (2C t ) were determined by comparing the PCR protein quantification.
cycle threshold (C t ) between groups, after normalising The liver samples (25 mg) were homogenised in 300 μl
to the housekeeping gene 36β4. The data are expressed of lysis buffer and centrifuged (1000 g, 4◦ C, 10 min).
as fold induction over the control group, which was Then, the homogenates were diluted 1:3 before protein
set to 100%. The sequences of the forward and reverse quantification. For inguinal adipose tissue, we homo-
primers were 5 -CATCTGCTGGCCTTCTCCAA-3 and genised 40 mg of tissue in 300 μl of the pH 7.4 lysis buffer
5 -ATCCAGGCTCTCTGGCTTCTG-3 for the leptin (20 mM Tris-HCl, 10 mM NaF, 1% NP40, 150 mM NaCl,
gene, and 5 -TGTTTGACAACGGCAGCATTT-3 and 5 mM EDTA, 0.1% SDS) containing protease inhibitors.
5 -CCGAGGCAACAGTTGGGTA-3 for 36β4. For these The homogenates also underwent an ultrasonic procedure


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
J Physiol 590.21 Maternal high fat diet programmes adrenal and thyroid function 5507

(2 × 10 s pulse on and 20 s pulse off ) followed by two Catecholamine quantification


subsequent centrifugations (9500 g, 4◦ C, 15 min). The
Total catecholamine (adrenaline and noradrenaline)
total protein content of the supernatants was quantified
content was quantified using a trihydroxyindole
using a BCA kit (Thermo Scientific). Both liver and
method with some modifications (Trevenzoli et al.
adipose tissue were used for analysis of AR content.
2007). We used 50 μl of the supernatant from the
All samples were denatured in the sample buffer (50 mM
homogenised glands to quantify the catecholamines.
Tris-HCl, pH 6.8, 1% SDS, 5% 2-mercaptoethanol, 10%
Adrenaline was used as standard. Briefly, 50 μl of the
glycerol, 0.001% bromophenol blue) and heated at 95◦ C
standard/supernatant/medium was mixed with 250 μl
for 5 min.
of 0.5 M phosphate buffer (pH 7.0) and 25 μl of 0.5%
Supernatants were analysed by SDS-PAGE, with
potassium ferricyanate; the mixing was followed by
a 10% or 12% polyacrylamide gel, and electro-
incubation (20 min; ice bath). The reaction was stopped
blotted onto PVDF membranes (Hybond P ECL
with 500 μl of 60 mg ml−1 ascorbic acid/5 N NaOH (1:19
membrane; Amersham Pharmacia Biotech, NJ, USA).
proportion). The parameters used in the fluorometer
The membranes were incubated with Tris-buffered
(Victor2, PerkinElmer, Turku, Finland) were 420 nm for
saline–Tween 20 (T-TBS) containing 5% non-fat dry
excitation and 510 nm for emission. The results were
milk for 90 min to block non-specific binding sites.
obtained by plotting the values as a linear regression of
Then, the membranes were washed with T-TBS and
the standard adrenaline curve. For this assay, we used nine
incubated overnight with specific primary antibodies.
adrenals from the C offspring and 10 adrenals from the
For the arcuate nucleus, anti-OBR (1:1000), anti-JAK2
HF offspring.
(1:500), anti-STAT3 (1:1000), anti-pSTAT3 (1:1000),
anti-suppressor of cytokine signalling 3 (SOCS3; 1:250)
(Santa Cruz Biotechnology, Santa Cruz, CA, USA) and
Liver glycogen content
anti-cyclophilin (1:3000) (Affinity BioReagents, IL, USA)
were used. For the PVN, anti-Pro TRH (1:500) (Life- Liver glycogen was measured using a protocol adapted
Span BioSciences, Seattle, WA, USA) and anti-cyclophilin from the Miller method (Trevenzoli et al. 2010b). Liver
(1:3000) were used. For adrenal gland, anti-TH (1:1000) samples were homogenised with 10% trichloroacetic acid
and anti-PNMT (1:1000) (Sigma-Aldrich, St Louis, MO, (100 mg of tissue/0.3 ml) and centrifuged (1000 g at 4◦ C
USA) were used. For adipose tissue, anti-β2AR (1:1000) for 10 min). To extract the glycogen, 200 μl of super-
(Sigma-Aldrich) and anti-β3AR (1:1000) (Santa Cruz natant was mixed with 667 μl of absolute ethanol. The
Biotechnology) were used. For liver, anti-β2AR (1:1000) mixture was maintained at 4◦ C for 24 h for glycogen
(Sigma-Aldrich) was used. Next, the membranes were precipitation and then centrifuged (1000 g at 4◦ C for
washed and incubated with the appropriate secondary 10 min); the resulting supernatant was then discarded.
antibodies, which were conjugated to biotin (Santa Cruz The glycogen pellets were hydrolysed with 0.1 ml of 1 M
Biotechnology; diluted in 0.5% non-fat dry milk/TBS) or HCl for 30 min at 100◦ C. Then, the mixtures were placed
horseradish peroxidase (HRP; Amersham, UK), for 1–2 h in an ice bath, and 0.1 ml of 1 M NaOH was added
at room temperature. The membranes that were incubated for neutralisation. The glucose produced by glycogen
in biotin-antibodies were washed and incubated for 1 h hydrolysis was measured using an enzymatic colorimetric
at room temperature with streptavidin HRP-conjugates method (Glucox commercial kit, Doles, Goiás, Brazil)
(Zymed, Camarillo CA, USA). Immunoreactive proteins and compared to a standard glucose curve. We used
were visualised using HRP substrate (ECL-plus; GE, Little nine C-offspring pups and 10 HF-offspring pups for this
Chalfont, Buckinghamshire, UK) and then exposed to analysis.
X-ray film. Finally, the area and density of the protein
bands were quantified using ImageJ 1.34s software (Wayne
Rasband, National Institutes of Health, Bethesda, MD, Statistical analysis
USA). All proteins were normalised by the cyclophilin
The data are expressed as the mean ± SEM. We used
content in each sample, except those from adipose
two-way ANOVA to analyse the body mass curve before
tissue, which were normalised by Ponceau staining (total
mating. The other variables were analysed using Student’s
protein).
unpaired t test. Results were considered significant when
For each protein analysed by Western blotting, we
P < 0.05.
used six or seven samples per group, and each sample
was obtained from a different animal. For analysis of
proteins in the hypothalamus, we used two animals
Results
(2 hypothalamic nuclei) to obtain each sample, and
five or six samples per group were used in the Wistar female rats fed with the high-fat diet did not show
experiments. increased body mass after 8 weeks of treatment (before


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
5508 J. G. Franco and others J Physiol 590.21

mating) compared to their controls (Fig. 1A). However, offspring. In addition, the HF offspring presented higher
the DEXA analysis showed that they had higher total body heart weights (ConOff: 0.190 ± 0.0087 g, and HFOff:
fat content (+27%, P < 0.05; Fig. 1B). At weaning, the 0.256 ± 0.006 g, P < 0.05). When analysed as a relative
control and HF mothers had similar body masses and fat proportion of body weight, the fat mass remained higher
contents (Fig. 1C and D). in the HF offspring (P < 0.05), but the heart weight was
Breast milk from the HF group contained more lactose similar between the groups (data not shown).
(+20%, P < 0.05) and triglycerides (+35%, P < 0.05) To verify whether hyperleptinaemia was associated with
and less cholesterol (−19%, P < 0.05) than the C group, alterations of central leptin signalling, we evaluated key
without any changes in protein content, on the 11th day proteins of the leptin pathway in the hypothalamic arcuate
of lactation (Fig. 2). At weaning (21st day of life), the nucleus. HF offspring had similar amounts of OBRb
breast milk had higher concentrations of proteins (+18%, and JAK2 compared with the controls (Fig. 5). However,
P < 0.05), cholesterol (+52%, P < 0.05) and triglycerides they had lower phosphorylated STAT3 (p-STAT3)/STAT3
(+86%, P < 0.05). The leptin concentration in the milk (−40%, P < 0.05; Fig. 5C), which is the main target
was similar between the two groups (Fig. 2). of the leptin pathway. This change was associated with
The maternal HF diet did not impact the birth reduced SOCS3 in the HF offspring (−56%, P < 0.05;
weight of the offspring (Fig. 3A). In contrast, at Fig. 5D).
weaning, the HF male offspring were evidently obese, The adrenal function evaluation showed that the
characterised by higher body mass (+53%, P < 0.05; HF offspring had higher adrenal catecholamine content
Fig. 3B) and increased fat mass in different adipose (+20%, P < 0.05; Fig. 6A) without changes in TH or
tissue depots: retroperitoneal (2.3-fold, P < 0.05; Fig. 4A), PNMT (Fig. 6C and D). Corticosteronaemia was similar
epididymal (3.4-fold, P < 0.05; Fig. 4A) and inguinal between the groups (Fig. 6B).
(2-fold, P < 0.05; Fig. 4A). This increased adiposity To evaluate the possible impact of the higher levels
was associated with higher leptin expression in the of catecholamines in HF offspring on other tissues, we
inguinal adipose tissue (2.5-fold, P < 0.05; Fig. 4B) and evaluated the expression of different adrenoceptors (ARs).
hyperleptinaemia (+62%, P < 0.05; Fig. 4C) in the HF HF offspring had less β3AR (−40%, P < 0.05; Fig. 7B),

Figure 1. Maternal body weight and adiposity


Body mass evolution and fat content/DEXA of control (filled bars; n = 6) and high-fat (open bars; n = 9) dams
before gestation (A and B, respectively), and body mass and fat content at weaning (C and D, respectively). Results
are expressed as means ± SEM; ∗ P < 0.05.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
J Physiol 590.21 Maternal high fat diet programmes adrenal and thyroid function 5509

without changes in β2AR (Fig. 7A), in the inguinal adipose serum TT3 (+15%, P < 0.05) and FT4 (+40%, P < 0.05)
tissue. without significant changes in TSH. This profile was
Despite the higher adiposity, which suggests insulin accompanied by increased pro-TRH in the PVN (+28%,
resistance, HF offspring presented hyperglycaemia P < 0.05).
(+30%, P < 0.05; Fig. 8A) and no alterations in serum
insulin, adiponectin or FFA (Fig. 8B–D).
HF offspring also presented higher absolute liver weight Discussion
(+46%, P < 0.05; Fig. 9A) and glycogen content (+50%,
P < 0.05; Fig. 9B). This alteration was accompanied by Normal leptinaemia in the perinatal period is an
reduced β2AR in the liver (−24%, P < 0.05; Fig. 9C). important factor for adequate development of the
When analysed as a relative proportion of body weight, central nervous system, especially the hypothalamus
liver weight trended toward being higher in the HF (Pinto et al. 2004; Bouret & Simerly, 2006), and also
offspring (P = 0.08; data not shown). for the physiology of peripheral tissues, such as adrenal
Figure 10 shows parameters of the hypothalamus– gland, liver, adipose tissue and thyroid, in rodents (Moura
pituitary–thyroid (HPT) axis. HF offspring had higher & Passos, 2005; Vickers et al. 2005). Both hyperleptinaemia

Figure 2. Breast milk composition


Milk content of protein (A), lactose (B), triglycerides (C), cholesterol (D) and leptin (E) of control (filled bars; n = 6)
and high-fat (open bars; n = 6) dams at the 11th day of lactation (middle lactation) and at weaning. Results are
expressed as means ± SEM; ∗ P < 0.05.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
5510 J. G. Franco and others J Physiol 590.21

(Toste et al. 2006b; Trevenzoli et al. 2010a,b) and


blockage of leptin action (Attig et al. 2008) during
lactation programme a cluster of metabolic dysfunctions
throughout life, including obesity and leptin resistance.
Maternal nutritional status may impact leptinaemia and
the metabolic parameters of the offspring with short- and
long-term consequences. Undernutrition during lactation
results in abolition of the leptin surge in the middle of
lactation and increased leptinaemia at weaning (Teixeira
et al. 2002). These changes are associated with central
resistance to the anorexigenic effect of leptin (Passos et al.
2004). However, the normalisation of leptin level pre-
vents the programming effects of undernutrition (Vickers
et al. 2005). In the present study, we found that a
maternal high-fat diet resulted in hyperleptinaemia in the
offspring at weaning, which was associated with higher
body weight and adiposity. These effects were independent
of maternal obesity at weaning, at which point the HF
mothers showed no change in body fat, suggesting a
specific effect of saturated fat on offspring metabolism.
The HF diet did markedly change the breast milk
composition, and the high concentrations of proteins,
lactose and triglycerides in the breast milk seemed
Figure 3. Effect of maternal HF diet on the offspring body
mass
to induce overnutrition in the offspring throughout
Birth weight (A) and body mass at weaning (B) of the control (C; lactation. Cunha et al. (2009) have shown that breast
n = 9) and high-fat (HF; n = 10) offspring. Results are expressed as milk from dams with pups that are raised in small
means ± SEM; ∗ P < 0.05.

Figure 4. Effect of maternal HF diet on the offspring adiposity and leptin production
Fat mass of retroperitoneal, epididymal and inguinal compartments (A), leptin RNAm expression in inguinal fat
pad (B) and leptinaemia (C) of the control (C; n = 9) and high-fat (HF; n = 10) offspring at weaning. Results are
expressed as means ± SEM; ∗ P < 0.05.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
J Physiol 590.21 Maternal high fat diet programmes adrenal and thyroid function 5511

litters (a recognised programming model of neonatal Our data show that even consumption of a normocaloric
overnutrition) also present high levels of triglycerides. A diet during pregnancy and lactation may cause deleterious
maternal HF diet increases the n3:n6 ratio and insulin effects in the offspring if the quality of the diet is not
level in breast milk from non-human primates without appropriate.
changing leptin content (Grant et al. 2011). Thus, our As a consequence of early overnutrition, the HF
results suggest that the precocious obesity programmed offspring presented increased visceral and subcutaneous
by a maternal HF diet is a consequence of changes in fat mass. Serum leptin level is positively correlated with
the macronutrients in breast milk rather than changes body weight and the amount of adipose tissue (Friedman
in the leptin concentration of milk that might disturb & Halaas, 1998), and the HF offspring presented hyper-
the leptin surge in the middle of lactation. Although leptinaemia associated with higher expression of leptin
many researchers are investigating the disturbances to in the inguinal adipose tissue. A similar phenotype was
the progeny caused by maternal malnutrition, most of observed in rats raised in small litters during lactation
them have used a drastic diet in terms of fat content (e.g. (Rodrigues et al. 2009).
50–60% of calories from fat), whereas the diet used in The anorexigenic effect of leptin is mediated
the present study was isocaloric and moderately high-fat. by the JAK2–STAT3 pathway in the hypothalamus.

Figure 5. Effect of maternal HF diet on leptin signalling pathway in the arcuate nucleus of the offspring
Content of OBRb (A), JAK2 (B), p-STAT3 (C) and SOCS3 (D) in samples of arcuate hypothalamic nuclei of control
(C; n = 6) and high-fat (HF; n = 6) offspring at weaning. Representative blots of leptin pathway proteins and
cyclophilin (control load) are shown (E). Results are expressed as means ± SEM; ∗ P < 0.05.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
5512 J. G. Franco and others J Physiol 590.21

When activated, p-STAT3 increases the synthesis of with important consequences, such as hyperphagia
anorexigenic neuropeptides (α-melanocyte-stimulating and decreased energy expenditure (Ahima & Lazar,
hormone (α-MSH) and cocaine and amphetamine 2008). In our experimental model, the analysis of the
regulated transcript (CART)) and reduces the production leptin pathway in the hypothalamic arcuate nucleus
of orexigenic factors (neuropeptide Y (NPY) and of the offspring showed that the maternal HF diet
agouti-related peptide (AgRP)). In rodents, these neural reduced STAT3 activation and SOCS3 content. These
pathways that regulate food intake and energy expenditure results indicate the beginning of leptin resistance, which
are not completely developed until the third postnatal may contribute to lower energy expenditure and the
week (Grove et al. 2005); however, in humans, they develop development of obesity. Because central feeding circuits
mainly prenatally (Djiane & Attig, 2008). In the present are still undeveloped during lactation (Grove & Smith,
study, dams were fed experimental diets until the end of 2003), we believe that leptin has a larger effect on
lactation, when the pups were 3 weeks old, specifically energy metabolism than on regulating food intake in this
to include this critical period of brain development in period.
rodents. Leptin resistance is frequently selective in obese humans
The JAK2–STAT3 pathway also stimulates the and animals. It is characterised by a lack of anorexigenic
transcription of SOCS3, a negative regulator of leptin and metabolic leptin responses and overactivation of
signalling (Bjorbaek et al. 1997, 1998). Hyperleptinaemia other systems influenced by this hormone, such as the
is frequently associated with central leptin resistance sympathetic nervous system (SNS).

Figure 6. Effect of maternal HF diet on adrenal function of the offspring


Adrenal content of catecholamines (A), serum corticosterone (B), adrenal content of tyrosine hydroxylase (TH;
C) and phenylethanolamine n-methyl transferase (PNMT; D) of control (C; n = 7–9) and high-fat (HF; n = 7–10)
offspring at weaning. Representative blots of catecholamine synthesising enzymes and cyclophilin (control load)
are shown (E). Results are expressed as means ± SEM; ∗ P < 0.05.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
J Physiol 590.21 Maternal high fat diet programmes adrenal and thyroid function 5513

The sympathoadrenal system is an important contribution of leptin action (Kalil & Haynes, 2012). We
component of physiological adaptation to challenging have shown that leptin treatment during the first 10 days of
situations. Changes in maternal diet, leptin levels or lactation increases catecholamine levels in the short and
nicotine exposure during lactation are stressful events, long term. These alterations are associated with higher
resulting in alterations in both the corticosterone and blood pressure and heart rate at adulthood (Trevenzoli
catecholamine levels of the offspring throughout life et al. 2007). In this study, we showed that a maternal
(Vieau et al. 2007; Fagundes et al. 2009; Trevenzoli et al. high-fat diet also increased adrenal catecholamine content
2010a; Laborie et al. 2011; Pinheiro et al. 2011). In in the pups at weaning, which may contribute to future
addition, these hormones make relevant contributions to programming of the cardiovascular system. Samuelsson
energy metabolism and cardiovascular function because et al. (2010) also demonstrated that maternal obesity
they affect glucose production and utilisation and fat induces hypertension development that is associated with
storage and protein metabolism. Thus, the HPA and SNS overactivity in 30-day-old rats. Thus, we suggest
sympathoadrenal system seems to be profoundly involved that adrenal catecholamines may contribute to this
with the developmental origins of obesity and cardio- phenotype.
vascular disease. The catecholamine synthesis pathway is regulated by
Leptin increases the sympathetic tone to brown TH and PNMT activities among other steps (Haycock &
adipose tissue, kidneys, hind limbs and adrenal glands. Wakade, 1992). We did not find differences in the content
Hypertension associated with obesity may have a great of these key enzymes between the groups that could have
explained the higher adrenal catecholamine content in the
HF offspring. However, the activities of TH and PNMT are
regulated by serine phosphorylation, and leptin directly
stimulates both TH expression and activity. This effect
is mediated by PKC and MAPK (Takekoshi et al. 2001;
Utsunomiya et al. 2001). The hyperleptinaemia found in
the HF offspring and/or the increased sympathetic tone to
the medulla may stimulate TH activity.
To investigate whether higher catecholamine
production in the HF offspring was associated with
tissue catecholamine effects, we measured ARs in tissues
that are profoundly regulated by these hormones.
We observed a lower content of β3AR in the adipose
tissue from the HF offspring, without an alteration in
β2AR. Lipolysis in white adipose tissue is stimulated by
catecholamines through βARs and the PKA pathway
that activates hormone-sensitive lipase (Langin, 2006).
Thus, our data suggest that the reduced lipolysis in the
HF offspring may have contributed to the development
of early obesity. Mice whose mothers were obese and
diabetic have lower ARβ2 and β3AR mRNA levels
and higher peroxisome proliferator-activated receptor
γ (PPARγ) mRNA levels in the inguinal fat tissue at
adulthood, suggesting decreased lipolysis and increased
lipogenesis (Samuelsson et al. 2010). It is possible
that high levels of catecholamines induce differential
downregulation of ARs. In addition, adipocytes are also
able to produce catecholamines, which might have an
important paracrine role on adiposity regulation and
deserves further investigation (Vargovic et al. 2011;
Kvetnansky et al. 2012).
Figure 7. Effect of maternal HF diet on content of Increased adiposity is strongly associated with insulin
adrenoreceptors in adipose tissue of the offspring resistance in humans and rodents. In fact, we observed
Content of β2 (A) and β3 (B) adrenoreceptors in the inguinal hyperglycaemia in our experimental model at weaning,
adipose tissue of control (C; n = 7) and high-fat (HF; n = 7) offspring
at weaning. Representative blots of ARs and Ponceau staining
suggesting glucose intolerance. However, serum insulin,
(control load) are shown (C). Results are expressed as means ± SEM; adiponectin and FFAs were unchanged in these animals.
∗ P < 0.05. We first tested the hypothesis that HF offspring would


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
5514 J. G. Franco and others J Physiol 590.21

have increased glycogenolysis induced by catecholamines, early adaptation in different experimental programming
by assessing the glycogen content and expression of β2-AR models (Toste et al. 2006a; Oliveira et al. 2009; Rodrigues
in the liver. HF offspring presented higher amounts et al. 2009). The HPT axis activity may be stimulated by
of glycogen with lower content of β2-AR, suggesting leptin under physiological conditions (Legradi et al. 1997;
reduced glycogenolysis. Thus, the hyperglycaemia found Nowak et al. 2002; Ortiga-Carvalho et al. 2002). Thus,
in the HF offspring seems not to be a consequence of we suppose that in our experimental model, the higher
increased glycogenolysis. On the other hand, a maternal levels of thyroid hormones in HF offspring occurred as
high-fat diet has been associated with an increased a consequence of hyperleptinaemia induced by obesity.
gluconeogenic capacity of offspring at birth through Hyperleptinaemia in obese humans (Pacifico et al. 2011)
epigenetic modifications, which could lead to excessive and rodents (Perello et al. 2010) is associated with
hepatic glucose production and altered insulin sensitivity high circulating thyroid hormones, suggesting that leptin
throughout life (Strakovsky et al. 2011). In addition, a still activates the HPT axis in these individuals even
maternal high-fat diet or maternal obesity can trigger though they are resistant to the anorexigenic effect of
liver lipotoxicity in rodents (Oben et al. 2010) and leptin.
non-human primates (McCurdy et al. 2009), which Despite having high levels of thyroid hormones, the
indicates liver insulin resistance and contributes to hepatic HF offspring were markedly obese, which seems counter-
glucose production. This phenotype at adulthood (high intuitive because thyroid hormones increase the metabolic
glycogen content and liver steatosis) may be associated rate and energy expenditure (Yen, 2001). However, in
with high leptin levels in the neonatal period, as we obese animals, these effects may be counterbalanced by an
have shown previously (Trevenzoli et al. 2010b). We also increase in food intake induced by T3 (Kong et al. 2004)
cannot discard the possibility of muscle insulin resistance and possibly by an impairment of the metabolic actions of
decreasing glucose uptake and contributing to the thyroid hormones, among other characteristics of obesity.
hyperglycaemia. Interestingly, a feature of the HF offspring was the small
Thyroid hormones classically have important roles but significant central activation of thyroid function, as
in early development. Our group have shown that suggested by the higher pro-TRH content in the PVN,
changes in thyroid function appear to be important for which may have caused the higher levels of T3 and T4.

Figure 8. Effect of maternal HF diet on glucose homeostasis of the offspring


Glycaemia (A), insulinaemia (B), adiponectinaemia (C) and serum free fatty acids (D) of control (C; n = 9) and
high-fat (HF; n = 10) offspring at weaning. Results are expressed as means ± SEM; ∗ P < 0.05.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
J Physiol 590.21 Maternal high fat diet programmes adrenal and thyroid function 5515

Figure 9. Effect of maternal HF diet on the liver content of glycogen and adrenoreceptor of the offspring
Liver weight (A), glycogen content (B) and content of β2 adrenoreceptor (C) in the liver of control (C; n = 7–9)
and high-fat (HF; n = 7–10) offspring at weaning. Representative blots of β2AR and cyclophilin (control load) are
shown (C). Results are expressed as means ± SEM; ∗ P < 0.05.

Figure 10. Effect of maternal HF diet on the thyroid function of the offspring
Serum total T3 (A), free T4 (B), TSH (C) and pro-TRH content in the hypothalamic paraventricular nucleus (D)
of control (C; n = 5–9) and high-fat (HF; n = 5–10) offspring at weaning. Representative blots of pro-TRH and
cyclophilin (control load) are shown (D). Results are expressed as means ± SEM; ∗ P < 0.05.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
5516 J. G. Franco and others J Physiol 590.21

These data are consistent with the results demonstrated by Djiane J & Attig L (2008). Role of leptin during perinatal
Perello et al. (2010) in DIO rats; they found a dissociation metabolic programming and obesity. J Physiol Pharmacol 59,
between leptin sensitivity in the arcuate and PVN hypo- 55–63.
thalamic nuclei, with a lack of anorexigenic response but Drummond GB (2009). Reporting ethical matters in The
preserved stimulation of TRH induced by leptin. Thus, Journal of Physiology: standards and advice. J Physiol 587,
713–719.
our data suggest that the hyperleptinaemia observed in
Fagundes AT, Moura EG, Passos MC, Santos-Silva AP, de
the HF offspring may have contributed to their higher Oliveira E, Trevenzoli IH, Casimiro-Lopes G, Nogueira-Neto
thyroid hormone levels. The normal serum TSH level JF & Lisboa PC (2009). Temporal evaluation of body
suggests a lower sensitivity of the pituitary to negative composition, glucose homeostasis and lipid profile of male
feedback from thyroid hormone; the HF offspring may rats programmed by maternal protein restriction during
also have had a new set point for regulation of the HPT lactation. Horm Metab Res 41, 866–873.
axis. Fischbeck KL & Rasmussen KM (1987). Effect of repeated
In the present study, we demonstrate for the first time reproductive cycles on maternal nutritional status,
that a perinatal maternal high-fat diet, through changes lactational performance and litter growth in ad libitum-fed
in milk composition, has short-term consequences, such and chronically food-restricted rats. J Nutr 117, 1967–1975.
as increased adrenal medullary and thyroid function, Friedman JM & Halaas JL (1998). Leptin and the regulation of
body weight in mammals. Nature 395, 763–770.
higher adiposity, hyperleptinaemia, and hyperglycaemia
Grant WF, Gillingham MB, Batra AK, Fewkes NM, Comstock
in the offspring. These changes occur even with a normal SM, Takahashi D, Braun TP, Grove KL, Friedman JE &
maternal caloric intake and may contribute, as priming Marks DL (2011). Maternal high fat diet is associated with
factors, to the development of metabolic and cardio- decreased plasma n-3 fatty acids and fetal hepatic apoptosis
vascular diseases. in nonhuman primates. PLoS One 6, e17261.
Grove KL, Grayson BE, Glavas MM, Xiao XQ & Smith MS
References (2005). Development of metabolic systems. Physiol Behav
86, 646–660.
Ahima RS & Lazar MA (2008). Adipokines and the peripheral Grove KL & Smith MS (2003). Ontogeny of the hypothalamic
and neural control of energy balance. Mol Endocrinol 22, neuropeptide Y system. Physiol Behav 79, 47–63.
1023–1031. Haycock JW & Wakade AR (1992). Activation and multiple-site
Attig L, Solomon G, Ferezou J, Abdennebi-Najar L, Taouis M, phosphorylation of tyrosine hydroxylase in perfused rat
Gertler A & Djiane J (2008). Early postnatal leptin blockage adrenal glands. J Neurochem 58, 57–64.
leads to a long-term leptin resistance and susceptibility to Helena CV, Szawka RE & Anselmo-Franci JA (2009).
diet-induced obesity in rats. Int J Obes (Lond) 32, Noradrenaline involvement in the negative-feedback effects
1153–1160. of ovarian steroids on luteinising hormone secretion.
Barker DJ, Eriksson JG, Forsen T & Osmond C (2002). Fetal J Neuroendocrinol 21, 805–812.
origins of adult disease: strength of effects and biological Kalil GZ & Haynes WG (2012). Sympathetic nervous system in
basis. Int J Epidemiol 31, 1235–1239. obesity-related hypertension: mechanisms and clinical
Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE & Flier JS implications. Hypertens Res 35, 4–16.
(1998). Identification of SOCS-3 as a potential mediator of Kim SY, Dietz PM, England L, Morrow B & Callaghan WM
central leptin resistance. Mol Cell 1, 619–625. (2007). Trends in pre-pregnancy obesity in nine states,
Bjorbaek C & Kahn BB (2004). Leptin signaling in the central 1993–2003. Obesity 15, 986–993.
nervous system and the periphery. Recent Prog Horm Res 59, Kong WM, Martin NM, Smith KL, Gardiner JV, Connoley IP,
305–331. Stephens DA, Dhillo WS, Ghatei MA, Small CJ & Bloom SR
Bjorbaek C, Uotani S, da Silva B & Flier JS (1997). Divergent (2004). Triiodothyronine stimulates food intake via the
signaling capacities of the long and short isoforms of the hypothalamic ventromedial nucleus independent
leptin receptor. J Biol Chem 272, 32686–32695. of changes in energy expenditure. Endocrinology 145,
Burdge GC, Hanson MA, Slater-Jefferies JL & Lillycrop KA 5252–5258.
(2007). Epigenetic regulation of transcription: a mechanism Kvetnansky R, Ukropec J, Laukova M, Manz B, Pacak K &
for inducing variations in phenotype (fetal programming) Vargovic P (2012). Stress stimulates production of
by differences in nutrition during early life? Br J Nutr 97, catecholamines in rat adipocytes. Cell Mol Neurobiol 32,
1036–1046. 801–813.
Bouret SG & Simerly RB (2006). Developmental programming Laborie C, Molendi-Coste O, Breton C, Montel V,
of hypothalamic feeding circuits. Clin Genet 70, 295–301. Vandenbulcke F, Grumolato L, Anouar Y & Vieau D (2011).
Callaway LK, Prins JB, Chang AM & McIntyre HD (2006). The Maternal perinatal undernutrition has long-term
prevalence and impact of overweight and obesity in an consequences on morphology, function and gene expression
Australian obstetric population. Med J Aust 184, 56–59. of the adrenal medulla in the adult male rat. J
Cunha AC, Pereira RO, Pereira MJ, Soares Vde M, Martins MR, Neuroendocrinol 23, 711–724.
Teixeira MT, Souza EP & Moura AS (2009). Long-term Langin D (2006). Control of fatty acid and glycerol release in
effects of overfeeding during lactation on insulin secretion – adipose tissue lipolysis. C R Biol 329, 598–607; discussion
the role of GLUT-2. J Nutr Biochem 20, 435–442. 653–655.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
J Physiol 590.21 Maternal high fat diet programmes adrenal and thyroid function 5517

Legradi G, Emerson CH, Ahima RS, Flier JS & Lechan RM Paxinos G & Watson C (2007). The Rat Brain in Stereotaxic
(1997). Leptin prevents fasting-induced suppression of Coordinates. Academic Press, New York, NY.
prothyrotropin-releasing hormone messenger ribonucleic Perello M, Cakir I, Cyr NE, Romero A, Stuart RC, Chiappini F,
acid in neurons of the hypothalamic paraventricular nucleus. Hollenberg AN & Nillni EA (2010). Maintenance of the
Endocrinology 138, 2569–2576. thyroid axis during diet-induced obesity in rodents is
Martin-Gronert MS & Ozanne SE (2010). Mechanisms linking controlled at the central level. Am J Physiol Endocrinol Metab
suboptimal early nutrition and increased risk of type 2 299, E976–989.
diabetes and obesity. J Nutr 140, 662–666. Pinheiro CR, Oliveira E, Trevenzoli IH, Manhaes AC,
Marques RG, Morales MM & Petroianu A (2009). Brazilian law Santos-Silva AP, Younes-Rapozo V, Claudio-Neto S, Santana
for scientific use of animals. Acta Cir Bras 24, 69–74. AC, Nascimento-Saba CC, Moura EG & Lisboa PC (2011).
McCurdy CE, Bishop JM, Williams SM, Grayson BE, Smith Developmental plasticity in adrenal function and leptin
MS, Friedman JE & Grove KL (2009). Maternal high-fat diet production primed by nicotine exposure during lactation:
triggers lipotoxicity in the fetal livers of nonhuman primates. gender differences in rats. Horm Metab Res 43,
J Clin Invest 119, 323–335. 693–701.
Moura EG, Lisboa PC & Passos MC (2008). Neonatal Pinto S, Roseberry AG, Liu H, Diano S, Shanabrough M, Cai X,
programming of neuroimmunomodulation – role of Friedman JM & Horvath TL (2004). Rapid rewiring of
adipocytokines and neuropeptides. arcuate nucleus feeding circuits by leptin. Science 304,
Neuroimmunomodulation 15, 176–188. 110–115.
Moura EG & Passos MC (2005). Neonatal programming of Reeves PG, Nielsen FH & Fahey GC Jr (1993). AIN-93 purified
body weight regulation and energetic metabolism. Biosci Rep diets for laboratory rodents: final report of the American
25, 251–269. Institute of Nutrition ad hoc writing committee on the
Nobre JL, Lisboa PC, Santos-Silva AP, Lima NS, Manhaes AC, reformulation of the AIN-76A rodent diet. J Nutr 123,
Nogueira-Neto JF, Cabanelas A, Pazos-Moura CC, Moura 1939–1951.
EG & de Oliveira E (2011). Calcium supplementation reverts Rodrigues AL, de Moura EG, Passos MC, Dutra SC & Lisboa
central adiposity, leptin, and insulin resistance in adult PC (2009). Postnatal early overnutrition changes the leptin
offspring programmed by neonatal nicotine exposure. J signalling pathway in the hypothalamic-pituitary-thyroid
Endocrinol 210, 349–359. axis of young and adult rats. J Physiol 587,
Nowak KW, Kaczmarek P, Mackowiak P, Ziolkowska A, 2647–2661.
Albertin G, Ginda WJ, Trejter M, Nussdorfer GG & Samuelsson AM, Matthews PA, Argenton M, Christie MR,
Malendowicz LK (2002). Rat thyroid gland expresses the McConnell JM, Jansen EH, Piersma AH, Ozanne SE, Twinn
long form of leptin receptors, and leptin stimulates the DF, Remacle C, Rowlerson A, Poston L & Taylor PD (2008).
function of the gland in euthyroid non-fasted animals. Int J Diet-induced obesity in female mice leads to offspring
Mol Med 9, 31–34. hyperphagia, adiposity, hypertension, and insulin resistance:
Oben JA, Mouralidarane A, Samuelsson AM, Matthews PJ, a novel murine model of developmental programming.
Morgan ML, McKee C, Soeda J, Fernandez-Twinn DS, Hypertension 51, 383–392.
Martin-Gronert MS, Ozanne SE, Sigala B, Novelli M, Poston Samuelsson AM, Morris A, Igosheva N, Kirk SL, Pombo JM,
L & Taylor PD (2010). Maternal obesity during pregnancy Coen CW, Poston L & Taylor PD (2010). Evidence for
and lactation programs the development of offspring sympathetic origins of hypertension in juvenile offspring of
non-alcoholic fatty liver disease in mice. J Hepatol 52, obese rats. Hypertension 55, 76–82.
913–920. Simmons R (2005). Developmental origins of adult metabolic
Oliveira E, Moura EG, Santos-Silva AP, Fagundes AT, Rios AS, disease: concepts and controversies. Trends Endocrinol Metab
Abreu-Villaca Y, Nogueira Neto JF, Passos MC & Lisboa PC 16, 390–394.
(2009). Short- and long-term effects of maternal nicotine Spencer SJ (2012). Early life programming of obesity: the
exposure during lactation on body adiposity, lipid profile, impact of the perinatal environment on the development of
and thyroid function of rat offspring. J Endocrinol 202, obesity and metabolic dysfunction in the offspring. Curr
397–405. Diabetes Rev 8, 55–68.
Ortiga-Carvalho TM, Oliveira KJ, Soares BA & Pazos-Moura Strakovsky RS, Zhang X, Zhou D & Pan YX (2011). Gestational
CC (2002). The role of leptin in the regulation of TSH high fat diet programs hepatic phosphoenolpyruvate
secretion in the fed state: in vivo and in vitro studies. carboxykinase gene expression and histone modification in
J Endocrinol 174, 121–125. neonatal offspring rats. J Physiol 589, 2707–2717.
Pacifico L, Anania C, Ferraro F, Andreoli GM & Chiesa C Sullivan EL, Smith MS & Grove KL (2011). Perinatal exposure
(2011). Thyroid function in childhood obesity and to high-fat diet programs energy balance, metabolism
metabolic comorbidity. Clin Chim Acta 413, 396–405. and behavior in adulthood. Neuroendocrinology 93,
Palkovits M (1973). Isolated removal of hypothalamic or other 1–8.
brain nuclei of the rat. Brain Res 59, 449–450. Takekoshi K, Ishii K, Nanmoku T, Shibuya S, Kawakami Y,
Passos MC, Vicente LL, Lisboa PC & de Moura EG (2004). Isobe K & Nakai T (2001). Leptin stimulates catecholamine
Absence of anorectic effect to acute peripheral leptin synthesis in a PKC-dependent manner in cultured porcine
treatment in adult rats whose mothers were malnourished adrenal medullary chromaffin cells. Endocrinology 142,
during lactation. Horm Metab Res 36, 625–629. 4861–4871.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society
5518 J. G. Franco and others J Physiol 590.21

Teixeira C, Passos M, Ramos C, Dutra S & Moura E (2002). Vargovic P, Ukropec J, Laukova M, Cleary S, Manz B,
Leptin serum concentration, food intake and body weight in Pacak K & Kvetnansky R (2011). Adipocytes as a new
rats whose mothers were exposed to malnutrition during source of catecholamine production. FEBS Lett 585,
lactation. J Nutr Biochem 13, 493. 2279–2284.
Toste FP, Alves SB, Dutra SC, Bonomo IT, Lisboa PC, Moura Vickers MH, Gluckman PD, Coveny AH, Hofman PL, Cutfield
EG & Passos MC (2006a). Temporal evaluation of the WS, Gertler A, Breier BH & Harris M (2005). Neonatal
thyroid function of rats programmed by leptin treatment on leptin treatment reverses developmental programming.
the neonatal period. Horm Metab Res 38, 827–831. Endocrinology 146, 4211–4216.
Toste FP, de Moura EG, Lisboa PC, Fagundes AT, de Oliveira E Vieau D, Sebaai N, Leonhardt M, Dutriez-Casteloot I,
& Passos MC (2006b). Neonatal leptin treatment Molendi-Coste O, Laborie C, Breton C, Deloof S & Lesage J
programmes leptin hypothalamic resistance and (2007). HPA axis programming by maternal undernutrition
intermediary metabolic parameters in adult rats. Br J Nutr in the male rat offspring. Psychoneuroendocrinology 32,
95, 830–837. 16–20.
Trevenzoli IH, Pinheiro CR, Conceicao EP, Oliveira E, Passos Yen PM (2001). Physiological and molecular basis of thyroid
MC, Lisboa PC & Moura EG (2010a). Programming of rat hormone action. Physiol Rev 81, 1097–1142.
adrenal medulla by neonatal hyperleptinemia: adrenal
morphology, catecholamine secretion, and leptin signaling
pathway. Am J Physiol Endocrinol Metab 298, E941–949. Author contributions
Trevenzoli IH, Rodrigues AL, Oliveira E, Thole AA, Carvalho L, Experiments were performed mainly in the Laboratory of
Figueiredo MS, Toste FP, Neto JF, Passos MC, Lisboa PC & Molecular Endocrinology of the Federal University of Rio de
Moura EG (2010b). Leptin treatment during lactation Janeiro and some procedures made in the Laboratory of End-
programs leptin synthesis, intermediate metabolism, and ocrine Physiology of the State University of Rio de Janeiro. J.G.F.,
liver microsteatosis in adult rats. Horm Metab Res 42,
T.P.F., C.P.D.R., C.M.C., and I.H.T. participated in collection,
483–490.
analysis and interpretation of data. J.G.F., C.C.P.-M., P.C.L.,
Trevenzoli IH, Valle MM, Machado FB, Garcia RM, Passos MC,
E.G.M., and I.H.T. performed drafting and critical revising of
Lisboa PC & Moura EG (2007). Neonatal hyperleptinaemia
programmes adrenal medullary function in adult rats: effects the manuscript. All authors have approved the final version of
on cardiovascular parameters. J Physiol 580, 629–637. the manuscript.
Troina AA, Figueiredo MS, Moura EG, Boaventura GT, Soares
LL, Cardozo LF, Oliveira E, Lisboa PC, Passos MA & Passos Acknowledgements
MC (2010). Maternal flaxseed diet during lactation alters
milk composition and programs the offspring body Research was supported by Conselho Nacional de
composition, lipid profile and sexual function. Food Chem Desenvolvimento Cientı́fico e Tecnológico (CNPq),
Toxicol 48, 697–703. Coordenação de Aperfeiçoamento de Pessoal de Nı́vel
Utsunomiya K, Yanagihara N, Tachikawa E, Cheah TB, Superior (CAPES) and Fundação de Amparo à Pesquisa do
Kajiwara K, Toyohira Y, Ueno S & Izumi F (2001). Rio de Janeiro (FAPERJ). T.P.F. was recipient of a FAPERJ
Stimulation of catecholamine synthesis in cultured bovine fellowship; C.C. was recipient of a CAPES fellowship and J.G.F.
adrenal medullary cells by leptin. J Neurochem 76, 926–934. was recipient of a CNP fellowship.


C 2012 The Authors. The Journal of Physiology 
C 2012 The Physiological Society

You might also like