SlideShare una empresa de Scribd logo
1 de 14
Descargar para leer sin conexión
Journal of Molecular and Cellular Cardiology 42 (2007) 526 – 539
www.elsevier.com/locate/yjmcc

Original article

NMR and cDNA array analysis prior to heart failure reveals an increase of
unsaturated lipids, a glutamine/glutamate ratio decrease and a specific
transcriptome adaptation in obese rat heart
J. Roncalli a , F. Smih a , F. Desmoulin b , N. Dumonteil a , R. Harmancey a , S. Hennig c , L. Perez a ,
A. Pathak a , M. Galinier a , P. Massabuau a , M. Malet-Martino b , J.M. Senard a , P. Rouet a,⁎
a

Unite de recherches sur les obesités, INSERM UPS U586, Institut Louis-Bugnard, Université Paul-Sabatier, CHU Rangueil,
BP 84225, 31432 Toulouse cedex 4, France
b
Groupe de RMN Biomédicale, Laboratoire SPCMIB (UMR CNRS 5068), Université Paul Sabatier, Toulouse, France
c
Deutsches Ressourcenzentrum für Genomforschung GmbH, Heubnerweg 6, D-14059 Berlin, Germany
Received 12 June 2006; received in revised form 13 October 2006; accepted 8 November 2006
Available online 11 January 2007

Abstract
Obesity is a risk factor for heart failure through a set of hemodynamic and hormonal adaptations, but its contribution at the molecular level is
not clearly known. Therefore, we investigated the kinetic cardiac transcriptome and metabolome in the Spontaneous Hypertensive Heart Failure
(SHHF) rat. The SHHF rat is devoid of leptin signaling when homozygous for a mutation of the leptin receptor (ObR) gene. The ObR−/− SHHF
rat is obese at 4 months of age and prone to heart failure after 14 months whereas its lean counterpart ObR−/+ is prone to heart failure after
16 months. We used a set of rat pangenomic high-density macroarrays to monitor left ventricle cardiac transcriptome regulation in 4- and 10month-old, lean and obese animals. Comparative analysis of left ventricle of 4- and 10-month-old lean rat revealed 222 differentially expressed
genes while 4- and 10-month-old obese rats showed 293 differentially expressed genes. 1H NMR analysis of the metabolome of left ventricular
extracts displayed a global decrease of metabolites, except for taurine, and lipid concentration. This may be attributed to gene expression
regulation and likely increased extracellular mass. The glutamine to glutamate ratio was significantly lower in the obese group. The relative
unsaturation of lipids increased in the obese heart; in particular, omega-3 lipid concentration was higher in the 10-month-old obese heart. Overall,
several specific kinetic molecular patterns act as a prelude to heart failure in the leptin signaling deficient SHHF obese rat.
© 2006 Elsevier Inc. All rights reserved.
Keywords: Obesity; Heart failure; NMR; Transcriptome; Metabolome; Arrays

1. Introduction
Obesity is defined by excessive adipose mass, which is
characterized by a body mass index (BMI) over 30 kg/m2.
Obesity prevalence is reaching around 10% to 30% of the
population in industrialized countries and this rate is doubling
every 10 years [1]. Obesity is leading to enhanced mortality [2]
and cardiovascular morbidity by means of arterial hypertension
(HTA) and endocrino-metabolic abnormalities like type II
⁎ Corresponding author. Faculté de Médecine, Laboratoire de Pharmacologie, INSERM U586, 37 allées Jules Guesde, 31073 Toulouse cedex 7, France.
Tel.: +33 5 61 14 59 98; fax: +33 5 61 25 51 16.
E-mail address: Philippe.Rouet@toulouse.inserm.fr (P. Rouet).
0022-2828/$ - see front matter © 2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.yjmcc.2006.11.007

diabetes or dyslipidemia, that are associated in the X metabolic
syndrome [3]. Obesity was shown to be an independent factor
for cardiovascular risk [4]. A positive correlation was noticed
between overweight and cardiovascular mortality, even after
adjusting for other risk factors. Additionally, obesity is an
independent risk factor for heart failure (HF): a BMI over
30 kg/m2 doubles the risk for HF in both genders [5].
Few works have studied specifically the cardiac abnormalities associated with obesity. Late stage obesity is characterized
by left ventricular hypertrophy (LVH), that leads to metabolic
change [6], and also by the alteration of the vegetative control of
cardiac frequency [7]. Severity and onset of these abnormalities
suggest a specific role for obesity, independent from HTA, in
the development of cardiomyopathies.
J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

Adipose tissue is now considered as an endocrine organ that
secretes a number of hormones and cytokines that are
regrouped under the name “adipokines” [8,9]. The hypothesis
of a specific role for adipose tissue has been supported by the
fact that many adipose tissue secretions have been shown to be
involved in cardiovascular pathologies [10]. We recently used a
dog nutritional model (hypercaloric high fat diet: HFD) of
obesity-related hypertension to show early-and late-specific
cardiac genes regulated at the transcriptome level [11,12].
These transcriptome regulations are responsible, at least in part,
for the cardiac remodeling that leads to LVH. More recently,
using human heart samples (right appendage) representative of
the right auricle obtained from non-diabetic patients undergoing open heart surgery and heart bypass, we have shown
different cardiac gene regulations that are specific to obesity
and independent from obesity-associated hypertension [13].
These observations are consistent with cardiac remodeling in
response to obesity which could be mediated, at least in part, by
adipokines.
In this work, our aim was to evaluate the molecular
mechanisms involved in the development of a dilated
cardiomyopathy (DCM) during the onset of heart failure (HF)
and the contribution of obesity in the early steps of the
development of this disease. Therefore, we used the SHHF/
Mcc-cp rat model of obesity, non-insulin dependent diabetes
and congestive heart failure [14]. As obese rats are prone to
heart failure several months before their lean counterpart [14],
experiments were performed with 4 and 10 months old, lean and
obese animals; before any sign of heart failure. We analyzed in
parallel the cardiac transcriptome and metabolome using a
cDNA array and an NMR approach respectively.
2. Materials and methods

527

After shaving of the chest, echocardiograms were performed
by using the Vivid 7 pro 7 echocardiographic system (GE
Medical System), equipped with a i13 L 14-MHz linear-array
transducer. Images were obtained from rats lightly anesthetized
by 1–2% isoflurane (AErrane, Baxter) lying on their back side
with transducer placed on the left hemithorax. Two-dimensional
parasternal long- and short-axis images of the left ventricle were
obtained, and two-dimensional targeted M-mode tracings were
recorded at a sweep speed of 200 mm/s. All measurements were
performed according to the recommendations of the American
Society for Echocardiography leading-edge method from three
consecutive cardiac cycles (n) with the roundness of the left
ventricular cavity (2D-image) as a criterion that the image was
on axis, great effort was taken to achieve a good image quality to
visualize the endocardial and epicardial borders of the heart by
gently moving and angulating the transducer. Measurements and
calculations used are as follows: percent LV fractional
shortening (FS), a measure of LV systolic function, was calculated as follows: FS = (EDD − ESD) / EDD × 100, where EDD
and ESD are end-diastolic and end-systolic diameters, respectively. All values were averaged over three consecutive cycles.
Twice a month, weight, systolic blood pressure (SBP) and
heart rate (HR) were measured in both lean and obese groups.
SBP and HR were recorded on vigil animals, after 15 min rest,
using a PowerLab System apparatus (ADInstruments, Australia). Attention was given to the possible appearance of CHF
symptoms such as subcutaneous oedema, hydrothorax, ascites,
dyspnea and cyanosis. At the end of the experiment, animals
were anesthetized, and surgery was quickly performed. Left
ventricular and right auricle samples were taken from 5 lean and
5 obese rats at the age of 4 months or 10 months. Samples were
immediately washed in cold Phosphate Buffer Saline Buffer,
snap frozen in liquid nitrogen and maintained at − 80 °C until
analysis.

2.1. Animals and general procedures
2.2. RNA extraction and cDNA labeling
All animal procedures were performed according to the
guidelines of the French Ministry of Agriculture. Rats were
housed at the Toulouse IFR31 animal facility in a room lit 12 h
per day (6 AM–6 PM) at an ambient temperature of 22 ± 1 °C.
Animals were allowed 1 week to adjust after arrival and had
access to regular rodent diet and tap water ad libitum during the
experiment.
Ten lean heterozygous (+/cp) and 10 obese homozygous
(cp/cp) Spontaneously Hypertensive and Heart Failure-prone
rats (SHHF), were provided by Charles River Laboratories
and included in the study. This strain was obtained by backcrossing a Koletsky obese rat to a Spontaneously Hypertensive Rat (SHR/N). Obese males develop congestive heart
failure (CHF) at 10–14 months, lean males at 14–18 months
[14,15]. From the Koletsky rat, SHHF rats harbor a nonsense
mutation of the leptin receptor gene, designated fa, resulting
in a premature stop codon in the leptin receptor. This mutation
is a recessive autosomic trait. As a consequence, cp/cp
homozygous animals for the fa mutation are obese. Lean rats
are heterozygous for this fa mutation or wild type for the cp
locus.

Total RNA was isolated from left ventricular samples; quality
check, concentration control and labeling were performed as
previously described [13].
2.3. cDNA array hybridization and expression analysis
cDNA array hybridization, washes, and expression analysis
were performed as previously described [13] using pangenomic macroarrays membranes (RZPD, Berlin) containing
26, 592 unique rat cDNA sequences spotted in duplicate. This
pangenomic array contained both known genes and nonannoted genes that required further computing analysis.
Therefore, to achieve the maximum information for the
respective probes on the membrane, the corresponding
accession numbers were used to identify both the respective
NCBI-Unigene clusters [16] and the respective EST contigs
provided by the TIGR gene index [17].
To limit the number of false positive results and to increase
specificity of our data, we used the following selection criteria
to determine whether the expression of a gene is up-regulated
528

J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

or down-regulated in a statistically significant manner: (1)
expression ratio of obese/lean was > 1.5 or <0.67, (2) expression
level was required to be over 3 times above membrane
background after local background subtraction (membrane
background was calculated by averaging the signal intensity
on empty spots i.e. generated by buffer deposition from DNA
free wells), (3) the difference between 2 experimental settings
was statistically significant by Student's t test, (4) attention was
given to avoid rejecting eventual highly induced genes with
initially low expression level (under 3 times the membrane
background). Statistical relevance of biological data and realtime PCR results was assessed with Student's t test or Mann–
Whitney rank sum test when normality test failed, using
SigmaStat 3.0 software (SPSS).
Hierarchical clustering was performed on normalized to the
mean intensity of all spots X-dot reader dataset with MultiExperiment Viewer 2.2 software (TIGR) [18]. The clustering
was performed on the average linkage clustering method for the
genes and the arrays.
2.4. Real-time PCR control of differential expression
A set of genes was randomly chosen for real-time PCR
validation of the observed differential expression [19]. Oligos
were synthesized by Proligo Company and designed with
Primer Express 2.0 software (supplemental Table 1 on-line).
Real time PCR was performed as previously described [11–13];
gene expressions were normalized to 18S RNA quantification
which has been found to be a reliable internal control gene in
our hands and others [20,21]. Real-time PCR data were
statistically analyzed with SigmaStat software (SPSS Science).
2.5. Heart left ventricular (LV) tissue dual phase metabolites
extraction
Frozen sections of LV (n = 20, 90 ± 39 mg) were powdered in
liquid nitrogen with a mortar and pestle and then immediately
subjected to a simultaneous extraction of lipids and watersoluble metabolites [22]. Briefly, frozen tissue fragments were
homogenized for 20 s in ice-cold purex-analytical-grade
methanol and chloroform in a 2:1 ratio (1.5 ml) by using an
Ultra-Turrax homogenizer. After 15 min in contact with the first
solvents at 4 °C, 0.5 ml chloroform and 0.5 ml distilled water
were added and homogenized to form an emulsion. The samples
were then centrifuged at 2000 × g for 30 min, 4 °C. The aqueous
phase was then separated from the organic phase. Both fractions
were dried in a Speed Vac concentrator. Extracts were maintained at − 80 °C until their preparation for NMR analysis. Prior
NMR analysis, extracts were reconstituted in 600 μl of D2O with
10 μl of a 10 mM 3-(trimethylsilyl)-1-propanesulfonate sodium
salt (TMPS) solution and 600 μl of CDCl3 / MeOD (2:1, v/v) for
aqueous fractions and organic fractions, respectively.
2.6. NMR analysis
1

H NMR spectra were recorded on a Bruker Avance 400
NMR spectrometer, equipped with a z-gradient 5 mm TBO

probe. Fully relaxed 1H spectra (without saturation effects)
were obtained in 12 min by accumulating 128 FID resulting
from 30° excitation pulses. Typical acquisition parameters were
a SW of 9.8 kHz, 32 K data points and 2.5 s repetition time. For
analysis of aqueous fractions, a presaturation pulse sequence
was used to suppress the residual intensity of the 1H water
resonance peak. Typical processing parameters were 65 K zerofilling and a LB of 1 Hz applied prior to Fourier transform.
Characteristic metabolites [23–25] and lipid signals [26–28]
have been assigned by reference to literature data and on the
basis of 2D homonuclear correlation spectroscopy (COSY)
experiments performed on the extracts (data not shown). The
signal of TMPS (δ = 0 ppm) for water-soluble metabolite and
residual CHCl3 (δ = 7.36 ppm) for lipids served as references
for chemical shift and concentration. A 32 mM concentration
of residual CHCl3 was previously determined with trichlorobenzene as standard for our batch of deuterated chloroform
[29]. Only resonances giving a signal to noise ratio over 15
were taken into account for statistical analysis. The number of
protons giving rise to a signal was considered in the
calculations of relative and absolute concentrations. Signals
to quantify water-soluble metabolites (chemical shift value in
ppm, and relative number of protons) were as fellows: lactate
(1.33, 3); alanine (1.48, 3) acetate (1.92, 3); glutamate (2.35,2);
glutamine (2.44, 2) creatine + phosphocreatine (3.04, 3), taurine
(3.41, 2; this resonance was chosen according to its specificity);
TMPS (0, 9); see also Fig. 3. Concentration of fatty acyl chains
was determined using the area of the α-methylene resonances
at 2.31 ppm as 100% of fatty acyl chains (peak 8 in Fig. 4).
Cholesterol was quantified using the area of the C-18 methyl
singlet at 0.67 ppm (peak 1 in Fig. 4). n-3 fatty acyl chains
were quantified using the characteristic triplet at 0.96 ppm of
the terminal-CH3 (peak 3 of Fig. 4). Total choline phospholipids were determined from the trimethyl group at 3.20 ppm
(peak 10 in Fig. 4). Mean unsaturation was calculated as the
ratio of vinyl from fatty chain (–CHfCH–) determined from
the signal at 5.35 ppm (peak 14 in Fig. 4) minus the
contribution of cholesterol proton, to the signal of total fatty
chains (peak 8 in Fig. 4). Mean poly-unsaturation was
calculated as the ratio of the signal of the allylic methylene
at 2.80 ppm (peak 9 in Fig. 4) to the signal of total fatty acyl
chains.
3. Results
3.1. Rat phenotype analysis
CHF symptoms such as subcutaneous oedema, hydrothorax,
ascites, dyspnea and cyanosis were not observed in lean and
obese 4- and 10-month-old animals. Four-month-old obese rats
had an average weight of 508 ± 12 g versus 386 ± 2 g for the lean
rats: this 32% increase in weight is statistically significant
(Student's t test, p = 0.008) (Table 1). At 10 months these
differences in weight were even more evident as obese rats had
an average weight of 727 ± 10 g and lean 477 ± 5 g; p ≤ 0.001).
Clearly, obesity was due to increased fat mass as reported for
this rat model [14].
J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

529

Table 1
Body and heart weight, blood pressure, BNP levels and cardiac echographic analysis
SHHF 4 months
Lean (n = 5)
Body weight (g)
Heart weight (g)
SBP (mm Hg)
BNP (ng/ml)
Diastolic septal wall thickness (mm)
Diastolic Posterior wall thickness (mm)
Systolic septal wall thickness (mm)
Systolic Posterior wall thickness (mm)
LV End diastolic diameter EDD (mm)
LV End systolic diameter ESD (mm)
LV Fraction shortening FS (%)

386 ± 2
1.27 ± 0.04
212 ± 7
3.8 ± 0.9
1.90 ± 0.16
2.15 ± 0.39
3.21 ± 0.18
3.41 ± 0.52
7.73 ± 0.61
4.30 ± 0.65
44.37 ± 6.70

SHHF 10 months
Obese (n = 5)

Lean (n = 5)

Obese (n = 5)

†

508 ± 12*
1.35 ± 0.02
204 ± 4
6.2 ± 2.2
1.78 ± 0.14
2.15 ± 0.34
3.18 ± 0.20
3.16 ± 0.37
7.78 ± 0.55
4.54 ± 0.49
41.65 ± 5.77

727 ± 10*,†
1.88 ± 0.09†
218 ± 2*,†
9.66 ± 2.3
2.18 ± 0.13*
2.32 ± 0.26
3.53 ± 0.26*
3.32 ± 0.27
8.56 ± 0.57*
5.20 ± 0.57*
39.25 ± 4.36

477 ± 5
1.7 ± 0.04†
207 ± 3
6.7 ± 2.8
2.13 ± 0.19*
2.08 ± 0.23
3.35 ± 0.25
3.2 ± 0.31
8.23 ± 0.48
4.87 ± 0.57
39.36 ± 7.26

Values statistically relevant are indicated by * for p < 0.05 (obese versus lean) and by † for 10-month-old versus 4-month-old statistical analysis (Student's t test). Data
are mean of five measurements per group ± S.E.M. Left ventricle (LV) percent left ventricular fractional shortening (FS) was calculated as follows: FS = (EDD − ESD) /
EDD × 100, where EDD and ESD are end-diastolic and end-systolic diameters, respectively. *p < 0.05 with Student's t test for obese or lean rats at 4-month-old versus
10-month-old.

Macroscopic examination of the heart did not reveal any
abnormalities or any cardiac infarction lesions. No significant
difference in weight were observed between lean and obese
average weight of the hearts in 4- or 10-month-old rats
(respectively 1.27 ± 0.04 g versus 1.35 ± 0.02 g; and 1.7 ±
0.04 g versus 1.88 ± 0.09 g). Nevertheless, heart mass significantly
increased in 10-month-old versus 4-month-old lean or obese rats.
Systolic blood pressure (SBP) was not statistically
significantly different: 212 ± 7 mm Hg in 4-month-old lean
rats versus 204 ± 4 mm Hg in obese rats. In 10-month-old
animals, SBP (207 ± 3 mm Hg) was maintained in lean rats
and was significantly increased in obese (218 ± 2 mm Hg).
Cardiac frequencies were similar in each group of animals
(data not shown). Moreover, we did not observe any clinical
sign of HF in 10-month-old animals (Table 1).
Echocardiographic analysis of obese SHHF and lean SHHF
animals demonstrated similar concentric LVH characterised by
increased wall thickness with similar LV cavity dimensions (Table
1). LV systolic function assessed by LV fractional shortening was
similar in both groups (Table 1) at 4 and 10 months. However, we
noted that in obese rats, there was an increase in diastolic and
systolic septal wall thickness and not in posterior wall thickness
between 4 and 10 months. End diastolic and systolic diameters
were also increased in obese rats. Moreover, echocardiographic
analysis did not reveal any left ventricular dysfunction.
3.2. Left ventricle transcriptome analysis
We first determined the number of detectable genes using
our cDNA macroarrays. Once the local background was
subtracted, we were able to detect an average of 9529 out of
26,592 genes with a signal greater than 1-fold and up to 37fold over the mean membrane background level (data not
shown). 7 out of 8 differentially expressed genes harboring
an expression level between 1.95 and 17.9 fold over
background were confirmed by Realtime PCR (87%
validation) (Table 2). According to this observation and previously published works [30], we defined an expression level

that had to be over 3 fold over background to be the minimum expression level for which we could reliably monitor
differential gene expression. The following data take this
parameter into account.
3.2.1. 10-month-old versus 4-month-old lean and obese rat
ventricle transcriptome analysis
Comparison of gene expression between 10-month-old
animals and 4-month-old animals revealed 222 differentially
Table 2
Comparison of DNA array analysis and real-time PCR
Gene name or GeneBank # Macroarray
Expr.
Ratio
level/bgd
Lean analysis
AA 858 801/NFKB1
AA963 792
AA 998 657
Obese analysis
AA 875 581/MRLCB
AI 028 924
AI 054 986
Gene common to lean and
obese analysis
AA 924 587
AA 819 584/ATP2A2

12.2
17
10

1.95
8.9
9.2

p

Ratio

p

10 m/4 m
10 m/4 m
2.59
0.0002 1.51
0.002
2.41
0.002 1.43
0.090
2.57
0.002 2.47
0.001
10 m/4 m
10 m/4 m
2.42
0.0014 2.26
0.017
2.65
0.017 2.46
0.003
2.87
0.01
2.16
0.018
10 m/4 m

4.36
17.9

rt-PCR

3.05
1.99

10 m/4 m
0.0005 1.63
0.005 2.09

0.038
0.038

Differential expression validation by realtime PCR for a set of randomly chosen
genes. Induction (if > 1.5) and repression (if < 0.6) ratios are represented. Mean
values are indicated for 5 measurements. cDNA macroarrays analysis were
performed on left ventricle (LV). m: months. Genes were randomly chosen
among those found to be statistically differentially regulated by Bioplot analysis
and harboring at least a 2 fold over the background expression intensity value.
Expr. level/bgd: ratio of signal intensity for a gene/mean background intensity.
Ratio Obese/Lean: signal intensity in Obese group/signal intensity in Lean
group. Ratio 10m/4m: signal intensity in 10-month-old group/signal intensity in
4-month-old group. NFKB1: nuclear factor kappa B p105 subunit; MRLCB:
myosin regulatory light chain; ATP2A2: ATPase, Ca2+ transporting, cardiac
muscle, slow twitch 2.
530

J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

clustering analysis, identity of 9 genes could be provided in the
main cluster (Fig. 2B and Table 4B).
3.3. Left ventricle metabolome analysis

Fig. 1. Differential expression analysis schematic diagram. Number of
differentially expressed genes is indicated. Arrows indicate the statistical
analysis of the data performed using Student's t test. 102 genes were found to be
common to the differentially expressed genes list from the obese and the lean
animals, 222 and 293 genes were found differentially expressed between 4- and
10-month-old lean and obese animals respectively.

expressed genes (17 repressed and 205 over-expressed) and 293
differentially expressed genes (30 repressed and 263 overexpressed) in lean and obese rats, respectively (Fig. 1).
Homologies searches generated a list of 95 defined genes
from the lean rat analysis and 132 defined genes from the obese
rat analysis (Table available as a supplementary data on line).
3.2.2. 10 months versus 4 months differential expression of a
set of cardiac-relevant genes
We identified a set of well-known function genes common to
lean and obese analysis (Table 3). These genes differentially
expressed between 4- and 10-month-old animals are indicators
of: neurohormonal activation (NPPA, ACE, ECE 1); apoptosis
(CASP1); inflammation (Il-6 receptor), fibrosis (TGF-β);
energetic metabolism (SLCA1, SLC25A10); hypertrophy
(GATA4); structure (MYH 6).
3.2.3. Hierarchical clustering of differentially expressed genes
Hierarchical clustering organization provided us with a
global view of the changes in cardiac gene expression induced
by the duration of hypertension in lean rats and the combination
of hypertension and obesity in obese rats. In addition,
hierarchical clustering was efficient at grouping gene expression profiles correctly without any intervention. Thus, gene
expression profiles were specific to the age of the lean or obese
SHHF rat hearts. Interpretation of the data is challenging for
some clusters due to a limited number of differentially
expressed genes, which limited cluster size. Nevertheless, one
could consider the set of 17 and 30 down-regulated genes in
10 months, lean (Fig. 2A) and obese (Fig. 2B) rats, respectively.
We first focused our attention on genes co-regulated in lean
animals and down regulated in 10-month-old animals (cluster 1,
Fig. 2A). We obtained gene identities or homologies for 11
genes out of 17 in this cluster (Table 4A). For obese animal

Characteristic signals arising from metabolites such as
creatine + phosphocreatine, taurine, glutamine, glutamate, acetate, alanine and lactate were detected in the 1H NMR spectrum
of the aqueous fraction of LV heart extract (Fig. 3).
Concentration of metabolites and statistical analysis of the
data showed a set of differences (Table 5). These differences
are related to the age and/or the obesity status. A significant
decrease in creatine and glutamate concentration was observed
and exclusively correlated to the age. Taurine concentration
was significantly decreased in the 10-month-old lean group
while its concentration was unchanged in other groups.
Glutamine concentration was significantly increased in the
lean group compared to that in the obese group. Moreover,
glutamine concentration was correlated to the age of rats and
significantly decreased in the 10-month-old lean group
compared to that in the 4-month-old lean group. The glutamine
to glutamate ratio was lower in obese animals, independent of
age.
A 1H NMR spectrum of total lipid extract of an obese SHHF
heart rat is shown in Fig. 4. Characteristic signals of lipid
moieties allowed the determination of total fatty acyl chains,
cholesterol, choline phospholipids (Table 6). Moreover, mean
unsaturation which represents the number of vinyl moieties per
fatty acyl chain and the mean poly-unsaturation which
represents the number of diallylic methylene per fatty acyl
chain were calculated. Choline phospholipids, cholesterol and
fatty acyl chains contents were similar at the same ages in
the obese group and in the lean group. Surprisingly, we

Table 3
Differentially expressed genes selection, according to their well-known
function, resulting from lean and obese SHHF 4-month-old/10-month-old
comparison
Gene name and Gene bank
accession #

Expr.
level/bgd

Ratio

p (Bioplot)

IL-6 R/AA 963 567
CASP 1/AI 071 441
GATA 4/AA 997 121
TGFB/AI 548 079
NPPA/AA 819 343
ACE/AI 556 575
ECE 1/AA 817 947
MYH 6/AA 819 464
FN 1/AA 955 600
SLC1A1/AA 996 752
SLC25A10/AA 859 666

4.26
9.27
3.51
3.26
3.04
5.68
3.21
4.87
14.2
5.31
3.15

1.74
2.14
2.38
1.64
1.67
1.66
2.11
0.58
1.88
2.03
0.56

0.006
0.038
0.027
0.033
0.0061
0.010
0.039
0.042
0.038
0.044
0.040

Expr. level/bgd: ratio of signal intensity for a gene/mean background intensity.
Ratio: signal intensity in 10-month-old group/signal intensity in 4-month-old
group. IL-6 R: InterLeukine 6 Receptor. CASP 1: Caspase 1. GATA 4: GATA
binding protein 4. TGFB: Transforming Growth Factor Beta. NPPA: Natriuretic
Peptide Precursor type A. ACE: Angiotensin Converting Enzyme. ECE 1:
Endothelin Converting Enzyme 1. MYH 6: Myosin Heavy chain polypeptide 6
(Myosin Heavy Chain alpha). FN 1: FibroNectine 1. SLC1A1: solute carrier
family 1, member 1. SLC25A10: solute carrier family 25, member 10.
J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

531

Fig. 2. Hierarchical cluster analysis of differential expression. (A) Clustering performed with lean transcriptome analysis data. (B) Clustering performed with obese
transcriptome analysis data. 1 to 5: 4-month-old animals; 6 to 10: 10-month-old animals. Genes close to each other harboring correlated expressions are illustrated by
the tree on the left side. Genes in cluster 1 from lean and obese analysis are detailed in Table 4.
532

J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

Fig. 2 (continued).
J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

533

Table 4
Genes identities for cluster 1 from Figs. 2A and B
#Gene ID

Gene name

Ratio

4 months

10 months

p-value

Identities and homologies

(A) Gene co-regulated in lean animals and down regulated in 10-month-old animals (cluster 1, Fig. 2A)
AA926219
B4GALT1
0.54
1.42
0.77
0.00025917
UDP-Gal:betaGlcNAc beta 1.4-galactosyltransferase.
Polypeptide 1 (Rattus norvegicus)
AI454214
0.59
1.54
0.91
0.04882843
No sequence homology
AA900596
CSN4
0.27
1.43
0.38
0.04986411
Similar to COP9 complex subunit 4 (Homo sapiens)
AA957598
0.27
1.68
0.46
0.03521052
No sequence homology
AI556930
PKD1
1.52
1.93
2.93
0.01708796
Polycystic kidney disease 1 (Pkd 1) and tuberous
sclerosis 2 (Tsc2) genes (Homo sapiens)
AI556150
0.66
1.49
0.98
0.04582136
No sequence homology
AA900578
ALAD
0.6
1.5
0.91
0.03495742
Aminolevulinate. delta-. dehydratase (Alad)
(Rattus norvegicus)
AA925164
0.63
1.47
0.93
0.00010271
No sequence homology
AI547418
0.64
1.85
1.19
8.90e − 05
Similar to XP_141317.3 RIKEN cDNA A430105I19
gene (Mus musculus)
AA925172
FGFR1OP
0.65
1.39
0.9
0.00086072
FGFR1 oncogene partner (Homo sapiens)
AA957860
0.66
1.49
0.98
0.00555646
No sequence homology
AI575762
CABP1
0.61
1.55
0.95
0.00496229
Calcium binding protein 1 (Cabp1) (Rattus norvegicus)
highly similar to NP_839983.1
sorting nexin 26 [Mus musculus]. Human trichohyalin.
Potential multiple
AI454874
THH
0.63
1.44
0.9
0.00654817
Roles as a functional EF-hand-like calcium-binding
protein. A cornified cell envelope precursor and an
intermediate filament-associated (cross-linking) protein
(Homo sapiens)
AI136136
CHMP1.5
0.62
1.55
0.95
0.00646823
CHMP1.5 protein (Homo sapiens) function: putative
vesicle trafficking with VPS4 and putative chromatin
structure regulation in the nuclear matrix
AI555253
0.62
1.54
0.96
6.78e − 05
Similar to KIAA1632 prot. proline–serine–threonine
phosphatase interacting protein 2 (Homo sapiens)
AA925236
0.64
1.61
1.03
0.00126788
No sequence homology
AA956865
SEB4D
0.66
1.6
1.06
0.0010949
Similar to dJ259A10.1 (ssDNA binding protein (SEB4D).
RNA-binding region (RNP1. RRM) (Homo sapiens)
(B) Genes co-regulated in obese animals and down regulated in 10-month-old animals (cluster 1, Fig. 2B)
AA859561
0.08
1.51
0.11
0.04220966
No sequence homology
AA900791
CFI
1.54
1.7
2.62
0.04129383
Complement factor I. (Rattus norvegicus)
AA925981
0.09
1.48
0.14
0.00609678
Similar to 2410001C21Rik protein (Homo sapiens)
chromosome 20 open reading frame 43
AI501277
0.52
1.45
0.75
0.00210187
No sequence homology
AI136302
0.55
1.57
0.86
0.00989101
No sequence homology
AA926219
B4GALT1
0.56
1.55
0.87
0.00531375
UDP-Gal:betaGlcNAc beta 1.4-galactosyltransferase.
polypeptide 1 (Rattus norvegicus)
AA925093
0.58
1.45
0.84
0.00311088
No sequence homology
AI043693
0.63
1.53
0.96
0.00859156
No sequence homology
AA925880
SPOCK2
0.58
2.16
1.25
0.00768181
Similar to testican-2 protein (LOC361840)
AA858560
0.49
1.79
0.88
0.00540406
No sequence homology
AA859429
0.59
1.63
0.96
0.01117883
Similar to hypothetical protein MGC31967(function:
translation initiation factor activity) (Rattus norvegicus)
AI145122
Centa2
0.62
1.65
1.03
0.03227733
Centaurin-alpha2 protein (Centa2) (Rattus norvegicus)
AI136080
0.63
1.73
1.09
0.037447
Similar to zinc finger protein 198
AI145732
0.6
1.8
1.08
0.01230905
No sequence homology
AA858454
0.65
1.71
1.12
0.02969404
Transcribed locus. Moderately similar to XP_346694.1
hypothetical gene supported by NM_022857
AI715210
KS1
0.64
1.5
0.96
0.0019953
KRAB/zinc finger suppressor protein 1
(KS1)(LOC246264)(Rattus norvegicus)
AI113019
0.57
2.21
1.25
0.00374868
No sequence homology
AI043993
0.62
1.45
0.9
0.00679676
No sequence homology
AI112905
0.65
2.22
1.45
0.00955169
No sequence homology
AI145673
0.65
1.97
1.28
0.00586599
Transcribed locus. Weakly similar to XP_346694.1
hypothetical gene supported by
NM_022857
AI030972
0.48
1.58
0.75
0.00063431
No sequence homology
(continued on next page)
534

J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

Table 4 (continued)
#Gene ID

Gene name

Ratio

4 months

10 months

p-value

Identities and homologies

(B) Genes co-regulated in obese animals and down regulated in 10-month-old animals (cluster 1, Fig. 2B)
AA900732
0.61
1.45
0.88
0.00367707
Transcribed locus. Similar to NP_080656.1
RIKEN cDNA 6530413N01 gene (Mus musculus)
AI060068
FABP3
0.62
1.44
0.9
0.00212252
Fatty acid binding protein 3 (Fabp3)
(Rattus norvegicus)
AA926237
TNNI3
0.66
2.06
1.36
0.01630635
Troponin 1. Type 3 (Tnni3) (Rattus norvegicus)
AI029660
0.52
1.56
0.82
0.00171603
No sequence homology
AA858808
0.63
2.09
1.32
0.0010659
No sequence homology
AA858866
NT5
0.6
1.81
1.09
0.00320482
5 Nucleotidase (Nt5) (Rattus norvegicus)
AA925124
KIR7.1
0.63
1.98
1.24
0.00392794
Inward rectifier potassium channel Kir7.1
[(Rattus norvegicus)
AI137849
0.66
1.57
1.04
0.01128975
No sequence homology

noticed a lower fatty acid chain concentration in 10-monthold animals when compared to the 4-month-old animals that
was significant for the obese group. Obese animals displayed
a higher level of n-3 fatty acid chains when compared to
their lean counterparts. In accordance with these observations, 10-month-old obese animals displayed enhanced
unsaturation and poly-unsaturation. Unsaturation levels
increased with age only in obese animals. Therefore,
increased unsaturation is related to the obesity state and its
duration.
4. Discussion
We combined cDNA macroarrays with NMR metabolic
profiling to characterize the cardiac transcriptome and metabolome of two groups of SHHF rats at 4 and 10 months of age

which differed in their state of obesity and the associated X
metabolic syndrome [14,15]. Our main objective was to define
the molecular adaptation in heart at the onset of heart failure
development and to evaluate the impact of obesity on the
mechanism of heart failure development. Prior to the molecular
adaptations analysis, we examined the biological parameters of
the subject animals. As previously described, the obese rats,
homozygous for the fa mutation, displayed a significant
increase of weight (+ 32% at 4 months; + 52% at 10 months;
Table 1).
However, we noticed two major distinctions when analyzing
the biological parameters. First, both groups of animals were
hypertensive and 10-month-old obese animals were significantly more hypertensive than their lean counterparts. This may be a
consequence of the obesity. Second, 10-month-old obese
animals were expected to be at the onset of a DCM [14] but

Fig. 3. 1H NMR spectrum of a water-soluble fraction of a heart extract from a lean SHHF rat. Within the aliphatic region (−0.2 to 4.5 ppm) of the NMR spectra,
resonances have been assigned to lactate (Lac), alanine (Ala), acetate (Acet), glutamate (Glu), glutamine (Gln), creatine + phosphocreatine (Cr + PCr), taurine (Taur),
Sodium 3-(trimethyl-silyl)-1-propanesuffonate (TMPS); pH = 7.4.
J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

535

Table 5
Quantitation of water-soluble metabolites by 1H NMR
SHHF 4 months
Lean
Taurine
Creatine + Phoshocreatine
Glutamate (Glu)
Glutamine (Gln)
Gln/Glu ratio
Acetate
Alanine
Lactate

SHHF 10 months
Obese

a

33.70 ± 3.55
16.55 ± 1.20 a
5.72 ± 0.56 a
7.82 ± 0.71 a
1.37 ± 0.12
1.70 ± 0.44
2.20 ± 0.12 a
14.49 ± 5.09

Lean

Obese

34.71 ± 5.98
15.24 ± 2.72 a
5.75 ± 0.93 a
6.56 ± 1.30 b
1.14 ± 0.10 b
1.62 ± 0.55
2.07 ± 0.05 b
21.45 ± 9.06 a

28.48 ± 3.32
12.19 ± 1.37
4.43 ± 0.76
6.71 ± 0.78
1.53 ± 0.23
1.71 ± 0.65
1.75 ± 0.32
11.03 ± 1.72

33.01 ± 3.52 b
10.98 ± 1.27
4.53 ± 0.83
5.47 ± 0.89 b
1.21 ± 0.09 b
1.14 ± 0.37
2.09 ± 0.44
10.26 ± 1.39

Data are means ± S.E. (in nmol/mg wet wt) from 5 hearts in each group.
a
Obese 4-month-old is significantly different from obese 10-month-old, or lean 4-month-old is significantly different from lean 10-month-old, Student's t test
p < 0.05.
b
Obese is significantly different from lean of the same age, Student's t test p < 0.05.

we did not observe any SBP normalization in this group of
animals. This is evidence for the lack of development of a DCM
in these 10-month-old animals. Nevertheless, our observations
are in accordance with previously published data [15]. Hearts
weights at 4 and 10 months of age were not significantly
different in the obese and lean groups. Thus, 10-month-old heart
weights were in accordance with published data [14] and
correspond to LVH hearts when compared to normotensive
Wistar–Kyoto or Sprague–Dawley rats of this age [14,31]. At
10 months of age we did not notice a more pronounced
macroscopic LVH in obese hearts when compared to lean
hearts. This lack of macroscopic differences was also reflected
in the BNP levels that were not significantly increased by

obesity in 10-month-old rats (Table 1). However, echocardiographic analysis displayed an increased diastolic and systolic
septal wall thickness in obese 10-month-old animals (Table 1).
This slight structural remodeling is likely a consequence of
blood volume increase observed in obese animals. Alternatively, it could be the result of altered kinetic gene regulations which
were observed between lean and obese animals, as discussed
below.
We confirmed differential expression of 2 fold above
background (Table 2 and data not shown), which is common
in microarray analysis. Therefore, to eliminate false positives
and focus on truly differentially expressed genes we set the limit
for reliable analysis of gene expression at 3-fold over

Fig. 4. 1H NMR spectrum of a total lipid extract from an obese SHHF heart rat. Main peaks or regions are assigned as follows: 1, Cholesterol (Chol, C18); 2, CH3
terminal of fatty acyl chain (FA) and Chol (C26, C27); 3, characteristic triplet of CH3 terminal of (n-3) polyunsaturated FA; 4, Chol (C19); 5, (CH2)n of FA; 6, –CH2–
CH2–COO–; 7, –CH2–CHfCH–CH2–CHfCH–CH2–; 8, –CH2–COO–; 9, –CHfCH–CH2–CHfCH–; 10, –N+(CH3)3 of phosphatidylcholine; 11, –CH2N+ of
phosphatidylcholine; 12, mainly glycerol (C1 and C3) and –O–CH2–CH2N+ of phosphatidylcholine; 13, esterified glycerol (C2); 14, –CHfCH– of FA and Chol
(C6); S, solvents (methanol + H2O).
536

J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

Table 6
Quantitation of lipids by NMR
SHHF 4 months
Lean
Acyl chains (nmol/mg w.w.)
(n-3) Acyl chains %
Cholesterol (nmol/mg w.w.)
Choline phospholipids (nmol/mg w.w.)
Mean unsaturation
Mean poly-unsaturation

103.49 ± 11.28
8.80 ± 0.56
5.09 ± 0.33
14.54 ± 1.00
1.70 ± 0.09
1.17 ± 0.06

SHHF 10 months
Obese
117.49 ± 16.87
10.35 ± 0.79 b
5.33 ± 0.95
15.72 ± 2.06 a
1.78 ± 0.12
1.23 ± 0.08 a

Lean
a

Obese

90.74 ± 6.11
8.11 ± 1.25
4.61 ± 0.62
13.57 ± 0.89
1.78 ± 0.10
1.23 ± 0.07

84.49 ± 4.73
9.52 ± 0.62 b
5.11 ± 0.78
12.72 ± 082
1.90 ± 0.02 b
1.33 ± 0.02 b

Data are means ± S.E. from 5 hearts in each group.
a
Obese 4 months is significantly different from obese 10 months, or lean 4-month-old is significantly different from lean 10-month-old, Student's t test p < 0.05.
b
Obese is significantly different from lean of the same age, Student's t test p < 0.05.

background. According to previously published work [30], this
lowers the false positive rate to 0.7%. According to these
criteria, we did not have any evidence of an effect of obesity in
4- or 10-month-old animals. However, macroarrays analysis on
a set of known cardiac genes recapitulated the differential
expression of genes involved in hypertension and the development of heart failure (Table 3).
Hierarchical clustering analysis of gene expression from lean
animals revealed in the same main cluster 3 genes out of 11
encoding proteasome proteins COP9 subunit 4 (CSN4),
aminolevulinate deltadehydratase (ALAD) and CHMP1.5
(Table 4A) PKD1 is a cation channel regulator [32], a complex
involved in many regulatory processes, including control of
development and regulation of morphogenesis. Two genes, UDPGal:betaGlcNAc beta 1,4-galactosyltransferase (B4GALT1) and
trichohyalin (THH), encode for proteins that have a structural
role. UDP N-acetylglucosamine β-1,4 galactosyltransferase is
a widely distributed enzyme which catalyzes the transfer of
galactose to N-acetylglucosamine residues of glycoproteins
and glycolipids [33]. Elevated B4GALT1 expression was
already shown to occur in the failing hearts of spontaneously
hypertensive rats [34] and was found down regulated in our
experiments that monitored rat heart gene expression prior to
HF. In this cluster of down regulated genes, we also observed a
member of a novel Ca2+-binding protein subfamily (CABP1),
that is a component of Ca2+-mediated cellular signal transduction in the heart [35]. We also noticed a down regulation of
genes involved in cell proliferation and/or protein synthesis in
muscle and heart tissue such as the RNA binding protein
(SEB4D) that was shown to be also down regulated in colon
cancer [36]. FGFR1OP, which protects cells from apoptosis,
was down regulated as well [37]. Cluster 1 in the obese
analysis contained a set of genes encoding remodeling and
structural proteins and a second group of genes with designated
miscellaneous functions (Table 4B). In the group of genes
encoding remodeling and structural proteins, we noticed upregulation of complement factor 1 (CF1). It is well known that
both free radicals and complement activation can injure tissue.
Local complement activation may represent a mechanism by
which free radicals mediate tissue injury [38]. It has been
shown that complement activation is directly involved in
chronically sustained myocardial damage [39]. Up-regulation
of CF1 probably contributes to cardiac remodeling. The second

gene is B4GALT1, mentioned above and the third gene is
SPOCK2. SPOCK2 encodes a protein called testican-2, which
is able to abrogate inhibition of membrane type metalloproteinases [40]. KS1 belongs to the largest family of zinc-finger
transcription factors containing the KRAB domain. The
functions proposed for members of the KRAB-containing
protein family are transcriptional repression of RNA polymerase and binding and splicing of RNA. KS1 counteracts
neoplastic transformation induced by several oncogenes [41].
Therefore KS1 could be involved in maintenance of the
nucleolus, cell differentiation, cell proliferation, apoptosis and
neoplastic transformation as found for other KRAB-containing
proteins [42]. In addition, we found heart-type fatty acid
binding protein (FABP3) down-regulated in this cluster.
FABP3 is involved in lipid metabolism and constitutes a
biochemical marker of myocyte injury in HF [43] whose
expression was found to be regulated in the heart in response to
fatty acid levels [44]. Interestingly, troponin (TNNI3), a well
known marker of HF [43], was also found down regulated in
this cluster. In the group of miscellaneous genes, we found
down regulation of 5′-nucleotidase (NT5). NT5 degrades the
adenosine moiety of ATP. Ischemic preconditioning was found
to activate NT5. Moreover, it was found that plasma adenosine
levels are increased in patients with chronic HF. NT5 activity
also increased in the blood and the myocardium in patients with
chronic HF, which may explain the increases in adenosine
levels in the plasma and the myocardium. In addition, it was
found that further elevation of plasma adenosine levels due to
either dipyridamole or dilazep reduces the severity of chronic
HF. Thus, endogenous adenosine was proposed for cardioprotection in chronic HF and against ischemia and reperfusion
injury [45]. The last gene in this cluster is KIR7.1, encoding for
an inwardly rectifying potassium (Kir) channel. Kir channels
are ubiquitously expressed and serve functions as diverse as
regulation of resting membrane potential, maintenance of K+
homeostasis, control of heart rate and hormone secretion [46].
NMR analysis allowed the quantitation of a number of
metabolites. Metabolite contents determined were in accordance with published data [47–49]. We noticed a general drop
in metabolite concentrations linked to the age of the animals. In
addition, the glutamine to glutamate ratio was significantly
lowered in obese animals. Globally lowered metabolite levels
can be explained by an increase of non-cellular mass such as
J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

excessive collagen type I and III that accumulate in the
myocardium in hypertensive heart tissue [50]. A lowered
glutamine to glutamate ratio could be a consequence of
increased TGF-β stimulation since the TGF-β upregulates the
phosphate dependant glutaminase and the Na+/H+ exchanger 3
gene expression pathway [51,52] favoring intracellular glutamine conversion to glutamate. Moreover, plasmatic TGF-β
circulating levels are increased in obesity [53] and favor
extracellular matrix expansion [54]. In addition, this lowered
glutamine to glutamate ratio in obese may reflect the use of
glutamine to generate more ATP to fuel the cardiomyocytes and
to respond to an increased energy need [52].
Fatty acyl chain concentrations were lower in 10-monthold animals when compared to 4-month-old obese animals. In
accordance with this observation, FABP3, involved in lipid
uptake, was down regulated in obese 10-month-old animals
versus 4-month-old (Table 4B). Besides, this apparent
lowering of fatty acyl chain concentrations might also be
related to the increase of myocardial mass by extracellular
matrix accumulation as mentioned above [55]. In accordance
with this hypothesis, we observed a kinetic up-regulation of
fribronectin gene expression in the obese group (supplemental
Table 2 on-line). Fibronectin is known to accumulate in the
interstitial space [55]. Since extracellular matrix accumulation
is a known occurrence in the remodelling heart and global
metabolite levels were lower in obese animals, (except for
taurine), this suggests that the obese heart is more fibrotic
than its lean counterpart at the same age. This would explain
earlier heart failure development in the obese compared to the
lean SHHF rat [14]. Interestingly, n-3 fatty acyl chains
percentage was higher in obese animals. This observation was
confirmed by an increased total fatty acyl unsaturation in
obese animals. Hypertension and obesity are known to induce
myocardium remodelling [56,57]. Such omega-3 accumulation has already been recently observed during muscle
regeneration [27] and may reflect mechanisms involved in
the increase of septal wall thickness as observed in the
10 months obese rats (Table 1). The increase of unsaturated
lipids was accompanied by conservation of taurine levels
(Table 5). Taurine was shown to preserve unsaturated
membrane lipids from lipid peroxidation [58]. We propose
that cardiac remodelling may share some similarities with
muscle regeneration. This increase of omega-3 may contribute
to the stimulation of membrane expansion from cardiac cells
as observed in PC12 cells [59].
In this present work we could identify a transcriptome kinetic
adaptation that was different in lean and obese animals (Figs. 1
and 2) but the 10-month-old lean and obese animals had a very
similar transcriptome. In our previous work performed on
human right appendage biopsies, we had shown that obese
hypertensive patients had clearly distinct cardiac gene expression patterns when compared to hypertensive patients. Thus,
several hypotheses are plausible. We could surmise that extreme
hypertension in the rat masks most obesity's contribution to
gene regulation. In addition, the duration of obesity is much
longer in patients than in rats and this point may be important to
allow for observation of the contribution of obesity to gene

537

regulations at the transcriptome level in hypertensive animals.
Although, leptin is one element among a number of adipokines
that are present in obese's bloodstream and that can regulate
heart gene expression, the lack of leptin signaling in the SHHF
obese rats may have some consequences on specific obesityinduced gene expression pattern. Indeed, it has been observed
that leptin added to cardiomyocytes in cell culture can induce
hypertrophy [60] and hyperplasia [61]. In hypertensive, nonobese humans, plasmatic levels of leptin have been associated
with myocardial wall thickness [62]. However, protein
synthesis regulation is known to occur not only at the mRNA
level and we did not perform a proteome analysis in this work to
investigate for post-transcriptional or post-traductional gene
regulations but rather a 1H NMR metabolomic analysis which
may be considered as a more downstream observation that takes
into account not only the protein level but also its enzymatic
activities. 1H NMR metabolomic analysis revealed significant
differences in left ventricle metabolites concentration between
obese and lean animals. Therefore, this work provides some
new insights both at the heart metabolome and transcriptome
level during the early steps of heart failure development in the
SHHF rat.
Acknowledgments
We thank Dr Sergueï Sokol (Toulouse Genomic Core
Facilities) respectively for array data WEB management. We
are indebted to Dr Peter J. Romanienko (Memorial SloanKettering Cancer Center, New York, New York, USA) for
critical reading of the manuscript.
Appendix A. Supplementary data
Supplementary data associated with this article can be found,
in the online version, at doi:10.1016/j.yjmcc.2006.11.007.
References
[1] Basdevant A. Obesity: epidemiology and public health. Ann Endocrinol
(Paris) 2000;61(Suppl 6):6–11.
[2] Contaldo F, Pasanisi F, Finelli C, de Simone G. Obesity, heart failure and
sudden death. Nutr Metab Cardiovasc Dis 2002;12:190–7.
[3] Mykkanen L, Kuusisto J, Pyorala K, Laakso M. Cardiovascular disease
risk factors as predictors of type 2 (non-insulin-dependent) diabetes
mellitus in elderly subjects. Diabetologia 1993;36:553–9.
[4] Hubert HB, Feinleib M, McNamara PM, Castelli WP. Obesity as an
independent risk factor for cardiovascular disease: a 26-year follow-up
of participants in the Framingham Heart Study. Circulation 1983;67:
968–77.
[5] Kenchaiah S, Evans JC, Levy D, Wilson PW, Benjamin EJ, Larson MG,
Kannel WB, Vasan RS. Obesity and the risk of heart failure. N Engl J Med
2002;347:305–13.
[6] Kagaya Y, Kanno Y, Takeyama D, Ishide N, Maruyama Y, Takahashi T,
Ido T, Takishima T. Effects of long-term pressure overload on regional
myocardial glucose and free fatty acid uptake in rats. A quantitative
autoradiographic study. Circulation 1990;81:1353–61.
[7] Beske SD, Alvarez GE, Ballard TP, Davy KP. Reduced cardiovagal
baroreflex gain in visceral obesity: implications for the metabolic
syndrome. Am J Physiol Heart Circ Physiol 2002;282:H630–5.
[8] Ahima RS, Flier JS. Adipose tissue as an endocrine organ. Trends
Endocrinol Metab 2000;11:327–32.
538

J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

[9] Guerre-Millo M. Adipose tissue and adipokines: for better or worse.
Diabetes Metab 2004;30:13–9.
[10] Guerre-Millo M. Adipose tissue hormones. J Endocrinol Invest
2002;25:855–61.
[11] Philip-Couderc P, Smih F, Pelat M, Vidal C, Verwaerde P, Pathak A, et al.
Cardiac transcriptome analysis in obesity-related hypertension.
Hypertension 2003;41:414–21.
[12] Philip-Couderc P, Smih F, Hall JE, Pathak A, Roncalli J, Harmancey R,
et al. Kinetic analysis of cardiac transcriptome regulation during chronic
high-fat diet in dogs. Physiol Genomics 2004;19:32–40.
[13] Philip-Couderc P, Pathak A, Smih F, Dambrin C, Harmancey R, Buys S,
et al. Uncomplicated human obesity is associated with a specific cardiac
transcriptome: involvement of the Wnt pathway. FASEB J 2004;18:
1539–40.
[14] McCune S, Baker P, Still H. SHHF/Mcc-cp rat: model of obesity, noninsulin-dependent diabetes, and congestive heart failure. ILAR News
1990;32:23–7.
[15] Heyen JR, Blasi ER, Nikula K, Rocha R, Daust HA, Frierdich G, et al.
Structural, functional, and molecular characterization of the SHHF model
of heart failure. Am J Physiol Heart Circ Physiol 2002;283:H1775–84.
[16] Wheeler DL, Church DM, Federhen S, Lash AE, Madden TL, Pontius JU,
et al. Database resources of the National Center for Biotechnology. Nucleic
Acids Res 2003;31:28–33.
[17] Liang F, Holt I, Pertea G, Karamycheva S, Salzberg SL, Quackenbush J.
Gene index analysis of the human genome estimates approximately
120,000 genes. Nat Genet 2000;25:239–40.
[18] Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, et al. TM4: a
free, open-source system for microarray data management and analysis.
BioTechniques 2003;34:374–8.
[19] Rajeevan MS, Ranamukhaarachchi DG, Vernon SD, Unger ER. Use of
real-time quantitative PCR to validate the results of cDNA array and
differential display PCR technologies. Methods 2001;25:443–51.
[20] Schmittgen TD, Zakrajsek BA. Effect of experimental treatment on
housekeeping gene expression: validation by real-time, quantitative RTPCR. J Biochem Biophys Methods 2000;46:69–81.
[21] Aerts JL, Gonzales MI, Topalian SL. Selection of appropriate control
genes to assess expression of tumor antigens using real-time RT-PCR.
BioTechniques 36 2004; 84–86, 88, 90–91.
[22] Tyagi RK, Azrad A, Degani H, Salomon Y. Simultaneous extraction of
cellular lipids and water-soluble metabolites: evaluation by NMR
spectroscopy. Magn Reson Med 1996;35:194–200.
[23] Desmoulin F, Confort-Gouny S, Masson S, Bernard M, Doddrell DM,
Cozzone PJ. Application of reverse-DEPT polarization transfer pulse
sequence to study the metabolism of carbon-13-labeled substrates in
perfused organs by 1H NMR spectroscopy. Magn Reson Med 1990;15:
456–61.
[24] Bollard ME, Murray AJ, Clarke K, Nicholson JK, Griffin JL. A study of
metabolic compartmentation in the rat heart and cardiac mitochondria
using high-resolution magic angle spinning 1H NMR spectroscopy. FEBS
Lett 2003;553:73–8.
[25] Mayr M, Metzler B, Chung YL, McGregor E, Mayr U, Troy H, et al.
Ischemic preconditioning exaggerates cardiac damage in PKC-delta null
mice. Am J Physiol Heart Circ Physiol 2004;287:H946–56.
[26] Adosraku RK, Choi GT, Constantinou-Kokotos V, Anderson MM,
Gibbons WA. NMR lipid profiles of cells, tissues, and body fluids:
proton NMR analysis of human erythrocyte lipids. J Lipid Res
1994;35:1925–31.
[27] Gillet B, Sebrie C, Bogaert A, Bleneau S, de la Porte S, Beloeil JC. Study
of muscle regeneration using in vitro 2D 1H spectroscopy. Biochim
Biophys Acta 2005;1724:333–44.
[28] Astrakas LG, Goljer I, Yasuhara S, Padfield KE, Zhang Q, Gopalan S, et al.
Proton NMR spectroscopy shows lipids accumulate in skeletal muscle in
response to burn trauma-induced apoptosis. FASEB J 2005;19:1431–40.
[29] Siddiqui N, Sim J, Silwood CJ, Toms H, Iles RA, Grootveld M.
Multicomponent analysis of encapsulated marine oil supplements using
high-resolution 1H and 13C NMR techniques. J Lipid Res 2003;44:
2406–27.
[30] Liu T, Lai H, Wu W, Chinn S, Wang PH. Developing a strategy to define

[31]

[32]

[33]
[34]

[35]

[36]

[37]

[38]

[39]

[40]

[41]

[42]
[43]
[44]

[45]

[46]
[47]

[48]

[49]

[50]

[51]

[52]
[53]

the effects of insulin-like growth factor-1 on gene expression profile in
cardiomyocytes. Circ Res 2001;88:1231–8.
Haas GJ, McCune SA, Brown DM, Cody RJ. Echocardiographic
characterization of left ventricular adaptation in a genetically determined
heart failure rat model. Am Heart J 1995;130:806–11.
Vandorpe DH, Chernova MN, Jiang L, Sellin LK, Wilhelm S, Stuart-Tilley
AK, et al. The cytoplasmic C-terminal fragment of polycystin-1 regulates a
Ca2+-permeable cation channel. J Biol Chem 2001;276:4093–101.
Shaper NL, Mann PL, Shaper JH. Cell surface galactosyltransferase:
immunochemical localization. J Cell Biochem 1985;28:229–39.
Humphries DE, Sirokman G, Bing OH. Enhanced galactosyltransferase
expression in the failing hearts of spontaneously hypertensive rats.
Biochem Biophys Res Commun 1996;218:320–4.
Haeseleer F, Sokal I, Verlinde CL, Erdjument-Bromage H, Tempst P,
Pronin AN, et al. Five members of a novel Ca(2+)-binding protein (CABP)
subfamily with similarity to calmodulin. J Biol Chem 2000;275:1247–60.
Scanlan MJ, Welt S, Gordon CM, Chen YT, Gure AO, Stockert E, et al.
Cancer-related serological recognition of human colon cancer:
identification of potential diagnostic and immunotherapeutic targets.
Cancer Res 2002;62:4041–7.
Guasch G, Ollendorff V, Borg JP, Birnbaum D, Pebusque MJ. 8p12 stem
cell myeloproliferative disorder: the FOP-fibroblast growth factor receptor
1 fusion protein of the t(6;8) translocation induces cell survival mediated
by mitogen-activated protein kinase and phosphatidylinositol 3-kinase/
Akt/mTOR pathways. Mol Cell Biol 2001;21:8129–42.
Tanhehco EJ, Yasojima K, McGeer PL, Washington RA, Lucchesi BR.
Free radicals upregulate complement expression in rabbit isolated heart.
Am J Physiol Heart Circ Physiol 2000;279:H195–201.
Yasojima K, Schwab C, McGeer EG, McGeer PL. Human heart generates
complement proteins that are upregulated and activated after myocardial
infarction. Circ Res 1998;83:860–9.
Nakada M, Miyamori H, Yamashita J, Sato H. Testican 2 abrogates
inhibition of membrane-type matrix metalloproteinases by other testican
family proteins. Cancer Res 2003;63:3364–9.
Gebelein B, Fernandez-Zapico M, Imoto M, Urrutia R. KRAB-independent
suppression of neoplastic cell growth by the novel zinc finger transcription
factor KS1. J Clin Invest 1998;102:1911–9.
Urrutia R. KRAB-containing zinc-finger repressor proteins. Genome Biol
2003;4:231.
Sato Y, Kita T, Takatsu Y, Kimura T. Biochemical markers of myocyte
injury in heart failure. Heart 2004;90:1110–3.
Van Bilsen M, de Vries JE, Van der Vusse GJ. Long-term effects of fatty
acids on cell viability and gene expression of neonatal cardiac myocytes.
Prostaglandins Leukot Essent Fatty Acids 1997;57:39–45.
Kitakaze M, Minamino T, Node K, Takashima S, Funaya H, Kuzuya T,
et al. Adenosine and cardioprotection in the diseased heart. Jpn Circ J
1999;63:231–43.
Abraham MR, Jahangir A, Alekseev AE, Terzic A. Channelopathies of
inwardly rectifying potassium channels. FASEB J 1999;13:1901–10.
Decker R, von Stuckrad-Barre S, Milakofsky L, Hofford JM, Harris N,
Vogel WH. Effect of stress on amino acids and related compounds in
various tissues of the rat. Life Sci 1995;57:1781–90.
Nielsen LB, Perko M, Arendrup H, Andersen CB. Microsomal triglyceride
transfer protein gene expression and triglyceride accumulation in hypoxic
human hearts. Arterioscler Thromb Vasc Biol 2002;22:1489–94.
Michael O'Donnell J, Narayan P, Bailey MQ, Abduljalil AM, Altschuld
RA, McCune SA, et al. 31P-NMR analysis of congestive heart failure in
the SHHF/Mcc-facp rat heart. J Mol Cell Cardiol 1998;30:235–41.
Diez J, Gonzalez A, Lopez B, Querejeta R. Mechanisms of disease:
pathologic structural remodeling is more than adaptive hypertrophy in
hypertensive heart disease. Nat Clin Pract Cardiovasc Med 2005;2:
209–16.
Nelson D, Rumsey WL, Erecinska M. Glutamine catabolism by heart
muscle. Properties of phosphate-activated glutaminase. Biochem J 1992
(Mar 1);282(Pt 2):559–64.
Welbourne T, Routh R, Yudkoff M, Nissim I. The glutamine/glutamate
couplet and cellular function. News Physiol Sci 2001;16:157–60.
Peterson MC. Circulating transforming growth factor beta-1: a partial
J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539

[54]
[55]

[56]

[57]

[58]

molecular explanation for associations between hypertension, diabetes,
obesity, smoking and human disease involving fibrosis. Med Sci Monit
2005;11:RA229–32.
Lijnen PJ, Petrov VV, Fagard RH. Induction of cardiac fibrosis by
transforming growth factor-beta(1). Mol Genet Metab 2000;71:418–35.
Sharov VG, Kostin S, Todor A, Schaper J, Sabbah HN. Expression of
cytoskeletal, linkage and extracellular proteins in failing dog myocardium.
Heart Fail Rev 2005;10:297–303.
Gosse P, Dallocchio M. Left ventricular hypertrophy: epidemiological
prognosis and associated critical factors. Eur Heart J 1993;14(Suppl
D):16–21.
Wakatsuki T, Schlessinger J, Elson EL. The biochemical response of the
heart to hypertension and exercise. Trends Biochem Sci 2004;29:
609–17.
Nandhini AT, Thirunavukkarasu V, Ravichandran MK, Anuradha CV.

[59]

[60]

[61]

[62]

539

Effect of taurine on biomarkers of oxidative stress in tissues of fructose-fed
insulin-resistant rats. Singapore Med J 2005;46:82–7.
Darios F, Davletov B. Omega-3 and omega-6 fatty acids stimulate
cell membrane expansion by acting on syntaxin 3. Nature 2006;440:
813–7.
Rajapurohitam V, Gan XT, Kirshenbaum LA, Karmazyn M. The obesityassociated peptide leptin induces hypertrophy in neonatal rat ventricular
myocytes. Circ Res 2003;93:277–9.
Tajmir P, Ceddia RB, Li RK, Coe IR, Sweeney G. Leptin increases
cardiomyocyte hyperplasia via extracellular signal-regulated kinase- and
phosphatidylinositol 3-kinase-dependent signaling pathways.
Endocrinology 2004;145:1550–5.
Paolisso G, Tagliamonte MR, Galderisi M, Zito GA, Petrocelli A, Carella
C, et al. Plasma leptin level is associated with myocardial wall thickness in
hypertensive insulin-resistant men. Hypertension 1999;34:1047–52.

Más contenido relacionado

Similar a C dna array analysis prior to heart failure reveals an incr

Heart Rhythm
Heart RhythmHeart Rhythm
Heart RhythmHany Abed
 
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...CrimsonPublishersIOD
 
An Unusual Case of Postpartum Thrombocytopenia
An Unusual Case of Postpartum ThrombocytopeniaAn Unusual Case of Postpartum Thrombocytopenia
An Unusual Case of Postpartum ThrombocytopeniaApollo Hospitals
 
Syndrome x eslam
Syndrome x eslamSyndrome x eslam
Syndrome x eslamEslam Habba
 
Endocrine cvs present copy
Endocrine  cvs present   copyEndocrine  cvs present   copy
Endocrine cvs present copyMahmoud Yossof
 
Functional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac diseaseFunctional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac diseaseLuis Carlos Murillo Valencia
 
Functional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the  fetus with non cardiac diseaseFunctional echocardiography in the  fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac diseasegisa_legal
 
Functional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac diseaseFunctional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac diseasegisa_legal
 
Potential role of uric acid in correlation with epidemics of hypertension and...
Potential role of uric acid in correlation with epidemics of hypertension and...Potential role of uric acid in correlation with epidemics of hypertension and...
Potential role of uric acid in correlation with epidemics of hypertension and...Apollo Hospitals
 
Presentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic SyndromePresentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic SyndromeJosh Nooner
 
Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome
Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome
Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome Josh Nooner
 
07 abdominal obesity_and_association_with.1
07   abdominal obesity_and_association_with.107   abdominal obesity_and_association_with.1
07 abdominal obesity_and_association_with.1Chistian Laguado
 
07 abdominal obesity_and_association_with.1
07   abdominal obesity_and_association_with.107   abdominal obesity_and_association_with.1
07 abdominal obesity_and_association_with.1Chistian Laguado
 
EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...
EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...
EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...oyepata
 

Similar a C dna array analysis prior to heart failure reveals an incr (20)

Heart Rhythm
Heart RhythmHeart Rhythm
Heart Rhythm
 
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
 
Journal of Research in Diabetes & Metabolism
Journal of Research in Diabetes & MetabolismJournal of Research in Diabetes & Metabolism
Journal of Research in Diabetes & Metabolism
 
An Unusual Case of Postpartum Thrombocytopenia
An Unusual Case of Postpartum ThrombocytopeniaAn Unusual Case of Postpartum Thrombocytopenia
An Unusual Case of Postpartum Thrombocytopenia
 
Syndrome x eslam
Syndrome x eslamSyndrome x eslam
Syndrome x eslam
 
CARDIO docx
CARDIO docxCARDIO docx
CARDIO docx
 
Adiponektin
AdiponektinAdiponektin
Adiponektin
 
Endocrine cvs present copy
Endocrine  cvs present   copyEndocrine  cvs present   copy
Endocrine cvs present copy
 
oski 4.pdf
oski 4.pdfoski 4.pdf
oski 4.pdf
 
Functional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac diseaseFunctional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac disease
 
Functional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the  fetus with non cardiac diseaseFunctional echocardiography in the  fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac disease
 
Functional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac diseaseFunctional echocardiography in the fetus with non cardiac disease
Functional echocardiography in the fetus with non cardiac disease
 
Potential role of uric acid in correlation with epidemics of hypertension and...
Potential role of uric acid in correlation with epidemics of hypertension and...Potential role of uric acid in correlation with epidemics of hypertension and...
Potential role of uric acid in correlation with epidemics of hypertension and...
 
Presentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic SyndromePresentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic Syndrome
 
Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome
Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome
Omega-3 Polyunsaturated Fatty Acids and Metabolic Syndrome
 
jf9020625-2
jf9020625-2jf9020625-2
jf9020625-2
 
Obesity ppt 2
Obesity ppt 2Obesity ppt 2
Obesity ppt 2
 
07 abdominal obesity_and_association_with.1
07   abdominal obesity_and_association_with.107   abdominal obesity_and_association_with.1
07 abdominal obesity_and_association_with.1
 
07 abdominal obesity_and_association_with.1
07   abdominal obesity_and_association_with.107   abdominal obesity_and_association_with.1
07 abdominal obesity_and_association_with.1
 
EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...
EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...
EFFECT OF ETHANOL LEAF EXTRACT OF Cassia angustifolia EXTRACT ON HEART AND LI...
 

Último

💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...Taniya Sharma
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.MiadAlsulami
 
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...narwatsonia7
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliRewAs ALI
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...narwatsonia7
 
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore EscortsVIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escortsaditipandeya
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...narwatsonia7
 
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Miss joya
 
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...Call girls in Ahmedabad High profile
 
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableVip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableNehru place Escorts
 
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls ServiceCALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls ServiceMiss joya
 
Call Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night Enjoy
Call Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night EnjoyCall Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night Enjoy
Call Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night Enjoynarwatsonia7
 
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...Miss joya
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service CoimbatoreCall Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatorenarwatsonia7
 
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiCall Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiNehru place Escorts
 
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...astropune
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girlsnehamumbai
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Miss joya
 

Último (20)

💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
 
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas Ali
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
 
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore EscortsVIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
 
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
 
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
 
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableVip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
 
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls ServiceCALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls Service
 
Call Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night Enjoy
Call Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night EnjoyCall Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night Enjoy
Call Girls Yelahanka Bangalore 📲 9907093804 💞 Full Night Enjoy
 
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCREscort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
 
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service CoimbatoreCall Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatore
 
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiCall Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
 
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
 

C dna array analysis prior to heart failure reveals an incr

  • 1. Journal of Molecular and Cellular Cardiology 42 (2007) 526 – 539 www.elsevier.com/locate/yjmcc Original article NMR and cDNA array analysis prior to heart failure reveals an increase of unsaturated lipids, a glutamine/glutamate ratio decrease and a specific transcriptome adaptation in obese rat heart J. Roncalli a , F. Smih a , F. Desmoulin b , N. Dumonteil a , R. Harmancey a , S. Hennig c , L. Perez a , A. Pathak a , M. Galinier a , P. Massabuau a , M. Malet-Martino b , J.M. Senard a , P. Rouet a,⁎ a Unite de recherches sur les obesités, INSERM UPS U586, Institut Louis-Bugnard, Université Paul-Sabatier, CHU Rangueil, BP 84225, 31432 Toulouse cedex 4, France b Groupe de RMN Biomédicale, Laboratoire SPCMIB (UMR CNRS 5068), Université Paul Sabatier, Toulouse, France c Deutsches Ressourcenzentrum für Genomforschung GmbH, Heubnerweg 6, D-14059 Berlin, Germany Received 12 June 2006; received in revised form 13 October 2006; accepted 8 November 2006 Available online 11 January 2007 Abstract Obesity is a risk factor for heart failure through a set of hemodynamic and hormonal adaptations, but its contribution at the molecular level is not clearly known. Therefore, we investigated the kinetic cardiac transcriptome and metabolome in the Spontaneous Hypertensive Heart Failure (SHHF) rat. The SHHF rat is devoid of leptin signaling when homozygous for a mutation of the leptin receptor (ObR) gene. The ObR−/− SHHF rat is obese at 4 months of age and prone to heart failure after 14 months whereas its lean counterpart ObR−/+ is prone to heart failure after 16 months. We used a set of rat pangenomic high-density macroarrays to monitor left ventricle cardiac transcriptome regulation in 4- and 10month-old, lean and obese animals. Comparative analysis of left ventricle of 4- and 10-month-old lean rat revealed 222 differentially expressed genes while 4- and 10-month-old obese rats showed 293 differentially expressed genes. 1H NMR analysis of the metabolome of left ventricular extracts displayed a global decrease of metabolites, except for taurine, and lipid concentration. This may be attributed to gene expression regulation and likely increased extracellular mass. The glutamine to glutamate ratio was significantly lower in the obese group. The relative unsaturation of lipids increased in the obese heart; in particular, omega-3 lipid concentration was higher in the 10-month-old obese heart. Overall, several specific kinetic molecular patterns act as a prelude to heart failure in the leptin signaling deficient SHHF obese rat. © 2006 Elsevier Inc. All rights reserved. Keywords: Obesity; Heart failure; NMR; Transcriptome; Metabolome; Arrays 1. Introduction Obesity is defined by excessive adipose mass, which is characterized by a body mass index (BMI) over 30 kg/m2. Obesity prevalence is reaching around 10% to 30% of the population in industrialized countries and this rate is doubling every 10 years [1]. Obesity is leading to enhanced mortality [2] and cardiovascular morbidity by means of arterial hypertension (HTA) and endocrino-metabolic abnormalities like type II ⁎ Corresponding author. Faculté de Médecine, Laboratoire de Pharmacologie, INSERM U586, 37 allées Jules Guesde, 31073 Toulouse cedex 7, France. Tel.: +33 5 61 14 59 98; fax: +33 5 61 25 51 16. E-mail address: Philippe.Rouet@toulouse.inserm.fr (P. Rouet). 0022-2828/$ - see front matter © 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.yjmcc.2006.11.007 diabetes or dyslipidemia, that are associated in the X metabolic syndrome [3]. Obesity was shown to be an independent factor for cardiovascular risk [4]. A positive correlation was noticed between overweight and cardiovascular mortality, even after adjusting for other risk factors. Additionally, obesity is an independent risk factor for heart failure (HF): a BMI over 30 kg/m2 doubles the risk for HF in both genders [5]. Few works have studied specifically the cardiac abnormalities associated with obesity. Late stage obesity is characterized by left ventricular hypertrophy (LVH), that leads to metabolic change [6], and also by the alteration of the vegetative control of cardiac frequency [7]. Severity and onset of these abnormalities suggest a specific role for obesity, independent from HTA, in the development of cardiomyopathies.
  • 2. J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 Adipose tissue is now considered as an endocrine organ that secretes a number of hormones and cytokines that are regrouped under the name “adipokines” [8,9]. The hypothesis of a specific role for adipose tissue has been supported by the fact that many adipose tissue secretions have been shown to be involved in cardiovascular pathologies [10]. We recently used a dog nutritional model (hypercaloric high fat diet: HFD) of obesity-related hypertension to show early-and late-specific cardiac genes regulated at the transcriptome level [11,12]. These transcriptome regulations are responsible, at least in part, for the cardiac remodeling that leads to LVH. More recently, using human heart samples (right appendage) representative of the right auricle obtained from non-diabetic patients undergoing open heart surgery and heart bypass, we have shown different cardiac gene regulations that are specific to obesity and independent from obesity-associated hypertension [13]. These observations are consistent with cardiac remodeling in response to obesity which could be mediated, at least in part, by adipokines. In this work, our aim was to evaluate the molecular mechanisms involved in the development of a dilated cardiomyopathy (DCM) during the onset of heart failure (HF) and the contribution of obesity in the early steps of the development of this disease. Therefore, we used the SHHF/ Mcc-cp rat model of obesity, non-insulin dependent diabetes and congestive heart failure [14]. As obese rats are prone to heart failure several months before their lean counterpart [14], experiments were performed with 4 and 10 months old, lean and obese animals; before any sign of heart failure. We analyzed in parallel the cardiac transcriptome and metabolome using a cDNA array and an NMR approach respectively. 2. Materials and methods 527 After shaving of the chest, echocardiograms were performed by using the Vivid 7 pro 7 echocardiographic system (GE Medical System), equipped with a i13 L 14-MHz linear-array transducer. Images were obtained from rats lightly anesthetized by 1–2% isoflurane (AErrane, Baxter) lying on their back side with transducer placed on the left hemithorax. Two-dimensional parasternal long- and short-axis images of the left ventricle were obtained, and two-dimensional targeted M-mode tracings were recorded at a sweep speed of 200 mm/s. All measurements were performed according to the recommendations of the American Society for Echocardiography leading-edge method from three consecutive cardiac cycles (n) with the roundness of the left ventricular cavity (2D-image) as a criterion that the image was on axis, great effort was taken to achieve a good image quality to visualize the endocardial and epicardial borders of the heart by gently moving and angulating the transducer. Measurements and calculations used are as follows: percent LV fractional shortening (FS), a measure of LV systolic function, was calculated as follows: FS = (EDD − ESD) / EDD × 100, where EDD and ESD are end-diastolic and end-systolic diameters, respectively. All values were averaged over three consecutive cycles. Twice a month, weight, systolic blood pressure (SBP) and heart rate (HR) were measured in both lean and obese groups. SBP and HR were recorded on vigil animals, after 15 min rest, using a PowerLab System apparatus (ADInstruments, Australia). Attention was given to the possible appearance of CHF symptoms such as subcutaneous oedema, hydrothorax, ascites, dyspnea and cyanosis. At the end of the experiment, animals were anesthetized, and surgery was quickly performed. Left ventricular and right auricle samples were taken from 5 lean and 5 obese rats at the age of 4 months or 10 months. Samples were immediately washed in cold Phosphate Buffer Saline Buffer, snap frozen in liquid nitrogen and maintained at − 80 °C until analysis. 2.1. Animals and general procedures 2.2. RNA extraction and cDNA labeling All animal procedures were performed according to the guidelines of the French Ministry of Agriculture. Rats were housed at the Toulouse IFR31 animal facility in a room lit 12 h per day (6 AM–6 PM) at an ambient temperature of 22 ± 1 °C. Animals were allowed 1 week to adjust after arrival and had access to regular rodent diet and tap water ad libitum during the experiment. Ten lean heterozygous (+/cp) and 10 obese homozygous (cp/cp) Spontaneously Hypertensive and Heart Failure-prone rats (SHHF), were provided by Charles River Laboratories and included in the study. This strain was obtained by backcrossing a Koletsky obese rat to a Spontaneously Hypertensive Rat (SHR/N). Obese males develop congestive heart failure (CHF) at 10–14 months, lean males at 14–18 months [14,15]. From the Koletsky rat, SHHF rats harbor a nonsense mutation of the leptin receptor gene, designated fa, resulting in a premature stop codon in the leptin receptor. This mutation is a recessive autosomic trait. As a consequence, cp/cp homozygous animals for the fa mutation are obese. Lean rats are heterozygous for this fa mutation or wild type for the cp locus. Total RNA was isolated from left ventricular samples; quality check, concentration control and labeling were performed as previously described [13]. 2.3. cDNA array hybridization and expression analysis cDNA array hybridization, washes, and expression analysis were performed as previously described [13] using pangenomic macroarrays membranes (RZPD, Berlin) containing 26, 592 unique rat cDNA sequences spotted in duplicate. This pangenomic array contained both known genes and nonannoted genes that required further computing analysis. Therefore, to achieve the maximum information for the respective probes on the membrane, the corresponding accession numbers were used to identify both the respective NCBI-Unigene clusters [16] and the respective EST contigs provided by the TIGR gene index [17]. To limit the number of false positive results and to increase specificity of our data, we used the following selection criteria to determine whether the expression of a gene is up-regulated
  • 3. 528 J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 or down-regulated in a statistically significant manner: (1) expression ratio of obese/lean was > 1.5 or <0.67, (2) expression level was required to be over 3 times above membrane background after local background subtraction (membrane background was calculated by averaging the signal intensity on empty spots i.e. generated by buffer deposition from DNA free wells), (3) the difference between 2 experimental settings was statistically significant by Student's t test, (4) attention was given to avoid rejecting eventual highly induced genes with initially low expression level (under 3 times the membrane background). Statistical relevance of biological data and realtime PCR results was assessed with Student's t test or Mann– Whitney rank sum test when normality test failed, using SigmaStat 3.0 software (SPSS). Hierarchical clustering was performed on normalized to the mean intensity of all spots X-dot reader dataset with MultiExperiment Viewer 2.2 software (TIGR) [18]. The clustering was performed on the average linkage clustering method for the genes and the arrays. 2.4. Real-time PCR control of differential expression A set of genes was randomly chosen for real-time PCR validation of the observed differential expression [19]. Oligos were synthesized by Proligo Company and designed with Primer Express 2.0 software (supplemental Table 1 on-line). Real time PCR was performed as previously described [11–13]; gene expressions were normalized to 18S RNA quantification which has been found to be a reliable internal control gene in our hands and others [20,21]. Real-time PCR data were statistically analyzed with SigmaStat software (SPSS Science). 2.5. Heart left ventricular (LV) tissue dual phase metabolites extraction Frozen sections of LV (n = 20, 90 ± 39 mg) were powdered in liquid nitrogen with a mortar and pestle and then immediately subjected to a simultaneous extraction of lipids and watersoluble metabolites [22]. Briefly, frozen tissue fragments were homogenized for 20 s in ice-cold purex-analytical-grade methanol and chloroform in a 2:1 ratio (1.5 ml) by using an Ultra-Turrax homogenizer. After 15 min in contact with the first solvents at 4 °C, 0.5 ml chloroform and 0.5 ml distilled water were added and homogenized to form an emulsion. The samples were then centrifuged at 2000 × g for 30 min, 4 °C. The aqueous phase was then separated from the organic phase. Both fractions were dried in a Speed Vac concentrator. Extracts were maintained at − 80 °C until their preparation for NMR analysis. Prior NMR analysis, extracts were reconstituted in 600 μl of D2O with 10 μl of a 10 mM 3-(trimethylsilyl)-1-propanesulfonate sodium salt (TMPS) solution and 600 μl of CDCl3 / MeOD (2:1, v/v) for aqueous fractions and organic fractions, respectively. 2.6. NMR analysis 1 H NMR spectra were recorded on a Bruker Avance 400 NMR spectrometer, equipped with a z-gradient 5 mm TBO probe. Fully relaxed 1H spectra (without saturation effects) were obtained in 12 min by accumulating 128 FID resulting from 30° excitation pulses. Typical acquisition parameters were a SW of 9.8 kHz, 32 K data points and 2.5 s repetition time. For analysis of aqueous fractions, a presaturation pulse sequence was used to suppress the residual intensity of the 1H water resonance peak. Typical processing parameters were 65 K zerofilling and a LB of 1 Hz applied prior to Fourier transform. Characteristic metabolites [23–25] and lipid signals [26–28] have been assigned by reference to literature data and on the basis of 2D homonuclear correlation spectroscopy (COSY) experiments performed on the extracts (data not shown). The signal of TMPS (δ = 0 ppm) for water-soluble metabolite and residual CHCl3 (δ = 7.36 ppm) for lipids served as references for chemical shift and concentration. A 32 mM concentration of residual CHCl3 was previously determined with trichlorobenzene as standard for our batch of deuterated chloroform [29]. Only resonances giving a signal to noise ratio over 15 were taken into account for statistical analysis. The number of protons giving rise to a signal was considered in the calculations of relative and absolute concentrations. Signals to quantify water-soluble metabolites (chemical shift value in ppm, and relative number of protons) were as fellows: lactate (1.33, 3); alanine (1.48, 3) acetate (1.92, 3); glutamate (2.35,2); glutamine (2.44, 2) creatine + phosphocreatine (3.04, 3), taurine (3.41, 2; this resonance was chosen according to its specificity); TMPS (0, 9); see also Fig. 3. Concentration of fatty acyl chains was determined using the area of the α-methylene resonances at 2.31 ppm as 100% of fatty acyl chains (peak 8 in Fig. 4). Cholesterol was quantified using the area of the C-18 methyl singlet at 0.67 ppm (peak 1 in Fig. 4). n-3 fatty acyl chains were quantified using the characteristic triplet at 0.96 ppm of the terminal-CH3 (peak 3 of Fig. 4). Total choline phospholipids were determined from the trimethyl group at 3.20 ppm (peak 10 in Fig. 4). Mean unsaturation was calculated as the ratio of vinyl from fatty chain (–CHfCH–) determined from the signal at 5.35 ppm (peak 14 in Fig. 4) minus the contribution of cholesterol proton, to the signal of total fatty chains (peak 8 in Fig. 4). Mean poly-unsaturation was calculated as the ratio of the signal of the allylic methylene at 2.80 ppm (peak 9 in Fig. 4) to the signal of total fatty acyl chains. 3. Results 3.1. Rat phenotype analysis CHF symptoms such as subcutaneous oedema, hydrothorax, ascites, dyspnea and cyanosis were not observed in lean and obese 4- and 10-month-old animals. Four-month-old obese rats had an average weight of 508 ± 12 g versus 386 ± 2 g for the lean rats: this 32% increase in weight is statistically significant (Student's t test, p = 0.008) (Table 1). At 10 months these differences in weight were even more evident as obese rats had an average weight of 727 ± 10 g and lean 477 ± 5 g; p ≤ 0.001). Clearly, obesity was due to increased fat mass as reported for this rat model [14].
  • 4. J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 529 Table 1 Body and heart weight, blood pressure, BNP levels and cardiac echographic analysis SHHF 4 months Lean (n = 5) Body weight (g) Heart weight (g) SBP (mm Hg) BNP (ng/ml) Diastolic septal wall thickness (mm) Diastolic Posterior wall thickness (mm) Systolic septal wall thickness (mm) Systolic Posterior wall thickness (mm) LV End diastolic diameter EDD (mm) LV End systolic diameter ESD (mm) LV Fraction shortening FS (%) 386 ± 2 1.27 ± 0.04 212 ± 7 3.8 ± 0.9 1.90 ± 0.16 2.15 ± 0.39 3.21 ± 0.18 3.41 ± 0.52 7.73 ± 0.61 4.30 ± 0.65 44.37 ± 6.70 SHHF 10 months Obese (n = 5) Lean (n = 5) Obese (n = 5) † 508 ± 12* 1.35 ± 0.02 204 ± 4 6.2 ± 2.2 1.78 ± 0.14 2.15 ± 0.34 3.18 ± 0.20 3.16 ± 0.37 7.78 ± 0.55 4.54 ± 0.49 41.65 ± 5.77 727 ± 10*,† 1.88 ± 0.09† 218 ± 2*,† 9.66 ± 2.3 2.18 ± 0.13* 2.32 ± 0.26 3.53 ± 0.26* 3.32 ± 0.27 8.56 ± 0.57* 5.20 ± 0.57* 39.25 ± 4.36 477 ± 5 1.7 ± 0.04† 207 ± 3 6.7 ± 2.8 2.13 ± 0.19* 2.08 ± 0.23 3.35 ± 0.25 3.2 ± 0.31 8.23 ± 0.48 4.87 ± 0.57 39.36 ± 7.26 Values statistically relevant are indicated by * for p < 0.05 (obese versus lean) and by † for 10-month-old versus 4-month-old statistical analysis (Student's t test). Data are mean of five measurements per group ± S.E.M. Left ventricle (LV) percent left ventricular fractional shortening (FS) was calculated as follows: FS = (EDD − ESD) / EDD × 100, where EDD and ESD are end-diastolic and end-systolic diameters, respectively. *p < 0.05 with Student's t test for obese or lean rats at 4-month-old versus 10-month-old. Macroscopic examination of the heart did not reveal any abnormalities or any cardiac infarction lesions. No significant difference in weight were observed between lean and obese average weight of the hearts in 4- or 10-month-old rats (respectively 1.27 ± 0.04 g versus 1.35 ± 0.02 g; and 1.7 ± 0.04 g versus 1.88 ± 0.09 g). Nevertheless, heart mass significantly increased in 10-month-old versus 4-month-old lean or obese rats. Systolic blood pressure (SBP) was not statistically significantly different: 212 ± 7 mm Hg in 4-month-old lean rats versus 204 ± 4 mm Hg in obese rats. In 10-month-old animals, SBP (207 ± 3 mm Hg) was maintained in lean rats and was significantly increased in obese (218 ± 2 mm Hg). Cardiac frequencies were similar in each group of animals (data not shown). Moreover, we did not observe any clinical sign of HF in 10-month-old animals (Table 1). Echocardiographic analysis of obese SHHF and lean SHHF animals demonstrated similar concentric LVH characterised by increased wall thickness with similar LV cavity dimensions (Table 1). LV systolic function assessed by LV fractional shortening was similar in both groups (Table 1) at 4 and 10 months. However, we noted that in obese rats, there was an increase in diastolic and systolic septal wall thickness and not in posterior wall thickness between 4 and 10 months. End diastolic and systolic diameters were also increased in obese rats. Moreover, echocardiographic analysis did not reveal any left ventricular dysfunction. 3.2. Left ventricle transcriptome analysis We first determined the number of detectable genes using our cDNA macroarrays. Once the local background was subtracted, we were able to detect an average of 9529 out of 26,592 genes with a signal greater than 1-fold and up to 37fold over the mean membrane background level (data not shown). 7 out of 8 differentially expressed genes harboring an expression level between 1.95 and 17.9 fold over background were confirmed by Realtime PCR (87% validation) (Table 2). According to this observation and previously published works [30], we defined an expression level that had to be over 3 fold over background to be the minimum expression level for which we could reliably monitor differential gene expression. The following data take this parameter into account. 3.2.1. 10-month-old versus 4-month-old lean and obese rat ventricle transcriptome analysis Comparison of gene expression between 10-month-old animals and 4-month-old animals revealed 222 differentially Table 2 Comparison of DNA array analysis and real-time PCR Gene name or GeneBank # Macroarray Expr. Ratio level/bgd Lean analysis AA 858 801/NFKB1 AA963 792 AA 998 657 Obese analysis AA 875 581/MRLCB AI 028 924 AI 054 986 Gene common to lean and obese analysis AA 924 587 AA 819 584/ATP2A2 12.2 17 10 1.95 8.9 9.2 p Ratio p 10 m/4 m 10 m/4 m 2.59 0.0002 1.51 0.002 2.41 0.002 1.43 0.090 2.57 0.002 2.47 0.001 10 m/4 m 10 m/4 m 2.42 0.0014 2.26 0.017 2.65 0.017 2.46 0.003 2.87 0.01 2.16 0.018 10 m/4 m 4.36 17.9 rt-PCR 3.05 1.99 10 m/4 m 0.0005 1.63 0.005 2.09 0.038 0.038 Differential expression validation by realtime PCR for a set of randomly chosen genes. Induction (if > 1.5) and repression (if < 0.6) ratios are represented. Mean values are indicated for 5 measurements. cDNA macroarrays analysis were performed on left ventricle (LV). m: months. Genes were randomly chosen among those found to be statistically differentially regulated by Bioplot analysis and harboring at least a 2 fold over the background expression intensity value. Expr. level/bgd: ratio of signal intensity for a gene/mean background intensity. Ratio Obese/Lean: signal intensity in Obese group/signal intensity in Lean group. Ratio 10m/4m: signal intensity in 10-month-old group/signal intensity in 4-month-old group. NFKB1: nuclear factor kappa B p105 subunit; MRLCB: myosin regulatory light chain; ATP2A2: ATPase, Ca2+ transporting, cardiac muscle, slow twitch 2.
  • 5. 530 J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 clustering analysis, identity of 9 genes could be provided in the main cluster (Fig. 2B and Table 4B). 3.3. Left ventricle metabolome analysis Fig. 1. Differential expression analysis schematic diagram. Number of differentially expressed genes is indicated. Arrows indicate the statistical analysis of the data performed using Student's t test. 102 genes were found to be common to the differentially expressed genes list from the obese and the lean animals, 222 and 293 genes were found differentially expressed between 4- and 10-month-old lean and obese animals respectively. expressed genes (17 repressed and 205 over-expressed) and 293 differentially expressed genes (30 repressed and 263 overexpressed) in lean and obese rats, respectively (Fig. 1). Homologies searches generated a list of 95 defined genes from the lean rat analysis and 132 defined genes from the obese rat analysis (Table available as a supplementary data on line). 3.2.2. 10 months versus 4 months differential expression of a set of cardiac-relevant genes We identified a set of well-known function genes common to lean and obese analysis (Table 3). These genes differentially expressed between 4- and 10-month-old animals are indicators of: neurohormonal activation (NPPA, ACE, ECE 1); apoptosis (CASP1); inflammation (Il-6 receptor), fibrosis (TGF-β); energetic metabolism (SLCA1, SLC25A10); hypertrophy (GATA4); structure (MYH 6). 3.2.3. Hierarchical clustering of differentially expressed genes Hierarchical clustering organization provided us with a global view of the changes in cardiac gene expression induced by the duration of hypertension in lean rats and the combination of hypertension and obesity in obese rats. In addition, hierarchical clustering was efficient at grouping gene expression profiles correctly without any intervention. Thus, gene expression profiles were specific to the age of the lean or obese SHHF rat hearts. Interpretation of the data is challenging for some clusters due to a limited number of differentially expressed genes, which limited cluster size. Nevertheless, one could consider the set of 17 and 30 down-regulated genes in 10 months, lean (Fig. 2A) and obese (Fig. 2B) rats, respectively. We first focused our attention on genes co-regulated in lean animals and down regulated in 10-month-old animals (cluster 1, Fig. 2A). We obtained gene identities or homologies for 11 genes out of 17 in this cluster (Table 4A). For obese animal Characteristic signals arising from metabolites such as creatine + phosphocreatine, taurine, glutamine, glutamate, acetate, alanine and lactate were detected in the 1H NMR spectrum of the aqueous fraction of LV heart extract (Fig. 3). Concentration of metabolites and statistical analysis of the data showed a set of differences (Table 5). These differences are related to the age and/or the obesity status. A significant decrease in creatine and glutamate concentration was observed and exclusively correlated to the age. Taurine concentration was significantly decreased in the 10-month-old lean group while its concentration was unchanged in other groups. Glutamine concentration was significantly increased in the lean group compared to that in the obese group. Moreover, glutamine concentration was correlated to the age of rats and significantly decreased in the 10-month-old lean group compared to that in the 4-month-old lean group. The glutamine to glutamate ratio was lower in obese animals, independent of age. A 1H NMR spectrum of total lipid extract of an obese SHHF heart rat is shown in Fig. 4. Characteristic signals of lipid moieties allowed the determination of total fatty acyl chains, cholesterol, choline phospholipids (Table 6). Moreover, mean unsaturation which represents the number of vinyl moieties per fatty acyl chain and the mean poly-unsaturation which represents the number of diallylic methylene per fatty acyl chain were calculated. Choline phospholipids, cholesterol and fatty acyl chains contents were similar at the same ages in the obese group and in the lean group. Surprisingly, we Table 3 Differentially expressed genes selection, according to their well-known function, resulting from lean and obese SHHF 4-month-old/10-month-old comparison Gene name and Gene bank accession # Expr. level/bgd Ratio p (Bioplot) IL-6 R/AA 963 567 CASP 1/AI 071 441 GATA 4/AA 997 121 TGFB/AI 548 079 NPPA/AA 819 343 ACE/AI 556 575 ECE 1/AA 817 947 MYH 6/AA 819 464 FN 1/AA 955 600 SLC1A1/AA 996 752 SLC25A10/AA 859 666 4.26 9.27 3.51 3.26 3.04 5.68 3.21 4.87 14.2 5.31 3.15 1.74 2.14 2.38 1.64 1.67 1.66 2.11 0.58 1.88 2.03 0.56 0.006 0.038 0.027 0.033 0.0061 0.010 0.039 0.042 0.038 0.044 0.040 Expr. level/bgd: ratio of signal intensity for a gene/mean background intensity. Ratio: signal intensity in 10-month-old group/signal intensity in 4-month-old group. IL-6 R: InterLeukine 6 Receptor. CASP 1: Caspase 1. GATA 4: GATA binding protein 4. TGFB: Transforming Growth Factor Beta. NPPA: Natriuretic Peptide Precursor type A. ACE: Angiotensin Converting Enzyme. ECE 1: Endothelin Converting Enzyme 1. MYH 6: Myosin Heavy chain polypeptide 6 (Myosin Heavy Chain alpha). FN 1: FibroNectine 1. SLC1A1: solute carrier family 1, member 1. SLC25A10: solute carrier family 25, member 10.
  • 6. J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 531 Fig. 2. Hierarchical cluster analysis of differential expression. (A) Clustering performed with lean transcriptome analysis data. (B) Clustering performed with obese transcriptome analysis data. 1 to 5: 4-month-old animals; 6 to 10: 10-month-old animals. Genes close to each other harboring correlated expressions are illustrated by the tree on the left side. Genes in cluster 1 from lean and obese analysis are detailed in Table 4.
  • 7. 532 J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 Fig. 2 (continued).
  • 8. J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 533 Table 4 Genes identities for cluster 1 from Figs. 2A and B #Gene ID Gene name Ratio 4 months 10 months p-value Identities and homologies (A) Gene co-regulated in lean animals and down regulated in 10-month-old animals (cluster 1, Fig. 2A) AA926219 B4GALT1 0.54 1.42 0.77 0.00025917 UDP-Gal:betaGlcNAc beta 1.4-galactosyltransferase. Polypeptide 1 (Rattus norvegicus) AI454214 0.59 1.54 0.91 0.04882843 No sequence homology AA900596 CSN4 0.27 1.43 0.38 0.04986411 Similar to COP9 complex subunit 4 (Homo sapiens) AA957598 0.27 1.68 0.46 0.03521052 No sequence homology AI556930 PKD1 1.52 1.93 2.93 0.01708796 Polycystic kidney disease 1 (Pkd 1) and tuberous sclerosis 2 (Tsc2) genes (Homo sapiens) AI556150 0.66 1.49 0.98 0.04582136 No sequence homology AA900578 ALAD 0.6 1.5 0.91 0.03495742 Aminolevulinate. delta-. dehydratase (Alad) (Rattus norvegicus) AA925164 0.63 1.47 0.93 0.00010271 No sequence homology AI547418 0.64 1.85 1.19 8.90e − 05 Similar to XP_141317.3 RIKEN cDNA A430105I19 gene (Mus musculus) AA925172 FGFR1OP 0.65 1.39 0.9 0.00086072 FGFR1 oncogene partner (Homo sapiens) AA957860 0.66 1.49 0.98 0.00555646 No sequence homology AI575762 CABP1 0.61 1.55 0.95 0.00496229 Calcium binding protein 1 (Cabp1) (Rattus norvegicus) highly similar to NP_839983.1 sorting nexin 26 [Mus musculus]. Human trichohyalin. Potential multiple AI454874 THH 0.63 1.44 0.9 0.00654817 Roles as a functional EF-hand-like calcium-binding protein. A cornified cell envelope precursor and an intermediate filament-associated (cross-linking) protein (Homo sapiens) AI136136 CHMP1.5 0.62 1.55 0.95 0.00646823 CHMP1.5 protein (Homo sapiens) function: putative vesicle trafficking with VPS4 and putative chromatin structure regulation in the nuclear matrix AI555253 0.62 1.54 0.96 6.78e − 05 Similar to KIAA1632 prot. proline–serine–threonine phosphatase interacting protein 2 (Homo sapiens) AA925236 0.64 1.61 1.03 0.00126788 No sequence homology AA956865 SEB4D 0.66 1.6 1.06 0.0010949 Similar to dJ259A10.1 (ssDNA binding protein (SEB4D). RNA-binding region (RNP1. RRM) (Homo sapiens) (B) Genes co-regulated in obese animals and down regulated in 10-month-old animals (cluster 1, Fig. 2B) AA859561 0.08 1.51 0.11 0.04220966 No sequence homology AA900791 CFI 1.54 1.7 2.62 0.04129383 Complement factor I. (Rattus norvegicus) AA925981 0.09 1.48 0.14 0.00609678 Similar to 2410001C21Rik protein (Homo sapiens) chromosome 20 open reading frame 43 AI501277 0.52 1.45 0.75 0.00210187 No sequence homology AI136302 0.55 1.57 0.86 0.00989101 No sequence homology AA926219 B4GALT1 0.56 1.55 0.87 0.00531375 UDP-Gal:betaGlcNAc beta 1.4-galactosyltransferase. polypeptide 1 (Rattus norvegicus) AA925093 0.58 1.45 0.84 0.00311088 No sequence homology AI043693 0.63 1.53 0.96 0.00859156 No sequence homology AA925880 SPOCK2 0.58 2.16 1.25 0.00768181 Similar to testican-2 protein (LOC361840) AA858560 0.49 1.79 0.88 0.00540406 No sequence homology AA859429 0.59 1.63 0.96 0.01117883 Similar to hypothetical protein MGC31967(function: translation initiation factor activity) (Rattus norvegicus) AI145122 Centa2 0.62 1.65 1.03 0.03227733 Centaurin-alpha2 protein (Centa2) (Rattus norvegicus) AI136080 0.63 1.73 1.09 0.037447 Similar to zinc finger protein 198 AI145732 0.6 1.8 1.08 0.01230905 No sequence homology AA858454 0.65 1.71 1.12 0.02969404 Transcribed locus. Moderately similar to XP_346694.1 hypothetical gene supported by NM_022857 AI715210 KS1 0.64 1.5 0.96 0.0019953 KRAB/zinc finger suppressor protein 1 (KS1)(LOC246264)(Rattus norvegicus) AI113019 0.57 2.21 1.25 0.00374868 No sequence homology AI043993 0.62 1.45 0.9 0.00679676 No sequence homology AI112905 0.65 2.22 1.45 0.00955169 No sequence homology AI145673 0.65 1.97 1.28 0.00586599 Transcribed locus. Weakly similar to XP_346694.1 hypothetical gene supported by NM_022857 AI030972 0.48 1.58 0.75 0.00063431 No sequence homology (continued on next page)
  • 9. 534 J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 Table 4 (continued) #Gene ID Gene name Ratio 4 months 10 months p-value Identities and homologies (B) Genes co-regulated in obese animals and down regulated in 10-month-old animals (cluster 1, Fig. 2B) AA900732 0.61 1.45 0.88 0.00367707 Transcribed locus. Similar to NP_080656.1 RIKEN cDNA 6530413N01 gene (Mus musculus) AI060068 FABP3 0.62 1.44 0.9 0.00212252 Fatty acid binding protein 3 (Fabp3) (Rattus norvegicus) AA926237 TNNI3 0.66 2.06 1.36 0.01630635 Troponin 1. Type 3 (Tnni3) (Rattus norvegicus) AI029660 0.52 1.56 0.82 0.00171603 No sequence homology AA858808 0.63 2.09 1.32 0.0010659 No sequence homology AA858866 NT5 0.6 1.81 1.09 0.00320482 5 Nucleotidase (Nt5) (Rattus norvegicus) AA925124 KIR7.1 0.63 1.98 1.24 0.00392794 Inward rectifier potassium channel Kir7.1 [(Rattus norvegicus) AI137849 0.66 1.57 1.04 0.01128975 No sequence homology noticed a lower fatty acid chain concentration in 10-monthold animals when compared to the 4-month-old animals that was significant for the obese group. Obese animals displayed a higher level of n-3 fatty acid chains when compared to their lean counterparts. In accordance with these observations, 10-month-old obese animals displayed enhanced unsaturation and poly-unsaturation. Unsaturation levels increased with age only in obese animals. Therefore, increased unsaturation is related to the obesity state and its duration. 4. Discussion We combined cDNA macroarrays with NMR metabolic profiling to characterize the cardiac transcriptome and metabolome of two groups of SHHF rats at 4 and 10 months of age which differed in their state of obesity and the associated X metabolic syndrome [14,15]. Our main objective was to define the molecular adaptation in heart at the onset of heart failure development and to evaluate the impact of obesity on the mechanism of heart failure development. Prior to the molecular adaptations analysis, we examined the biological parameters of the subject animals. As previously described, the obese rats, homozygous for the fa mutation, displayed a significant increase of weight (+ 32% at 4 months; + 52% at 10 months; Table 1). However, we noticed two major distinctions when analyzing the biological parameters. First, both groups of animals were hypertensive and 10-month-old obese animals were significantly more hypertensive than their lean counterparts. This may be a consequence of the obesity. Second, 10-month-old obese animals were expected to be at the onset of a DCM [14] but Fig. 3. 1H NMR spectrum of a water-soluble fraction of a heart extract from a lean SHHF rat. Within the aliphatic region (−0.2 to 4.5 ppm) of the NMR spectra, resonances have been assigned to lactate (Lac), alanine (Ala), acetate (Acet), glutamate (Glu), glutamine (Gln), creatine + phosphocreatine (Cr + PCr), taurine (Taur), Sodium 3-(trimethyl-silyl)-1-propanesuffonate (TMPS); pH = 7.4.
  • 10. J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 535 Table 5 Quantitation of water-soluble metabolites by 1H NMR SHHF 4 months Lean Taurine Creatine + Phoshocreatine Glutamate (Glu) Glutamine (Gln) Gln/Glu ratio Acetate Alanine Lactate SHHF 10 months Obese a 33.70 ± 3.55 16.55 ± 1.20 a 5.72 ± 0.56 a 7.82 ± 0.71 a 1.37 ± 0.12 1.70 ± 0.44 2.20 ± 0.12 a 14.49 ± 5.09 Lean Obese 34.71 ± 5.98 15.24 ± 2.72 a 5.75 ± 0.93 a 6.56 ± 1.30 b 1.14 ± 0.10 b 1.62 ± 0.55 2.07 ± 0.05 b 21.45 ± 9.06 a 28.48 ± 3.32 12.19 ± 1.37 4.43 ± 0.76 6.71 ± 0.78 1.53 ± 0.23 1.71 ± 0.65 1.75 ± 0.32 11.03 ± 1.72 33.01 ± 3.52 b 10.98 ± 1.27 4.53 ± 0.83 5.47 ± 0.89 b 1.21 ± 0.09 b 1.14 ± 0.37 2.09 ± 0.44 10.26 ± 1.39 Data are means ± S.E. (in nmol/mg wet wt) from 5 hearts in each group. a Obese 4-month-old is significantly different from obese 10-month-old, or lean 4-month-old is significantly different from lean 10-month-old, Student's t test p < 0.05. b Obese is significantly different from lean of the same age, Student's t test p < 0.05. we did not observe any SBP normalization in this group of animals. This is evidence for the lack of development of a DCM in these 10-month-old animals. Nevertheless, our observations are in accordance with previously published data [15]. Hearts weights at 4 and 10 months of age were not significantly different in the obese and lean groups. Thus, 10-month-old heart weights were in accordance with published data [14] and correspond to LVH hearts when compared to normotensive Wistar–Kyoto or Sprague–Dawley rats of this age [14,31]. At 10 months of age we did not notice a more pronounced macroscopic LVH in obese hearts when compared to lean hearts. This lack of macroscopic differences was also reflected in the BNP levels that were not significantly increased by obesity in 10-month-old rats (Table 1). However, echocardiographic analysis displayed an increased diastolic and systolic septal wall thickness in obese 10-month-old animals (Table 1). This slight structural remodeling is likely a consequence of blood volume increase observed in obese animals. Alternatively, it could be the result of altered kinetic gene regulations which were observed between lean and obese animals, as discussed below. We confirmed differential expression of 2 fold above background (Table 2 and data not shown), which is common in microarray analysis. Therefore, to eliminate false positives and focus on truly differentially expressed genes we set the limit for reliable analysis of gene expression at 3-fold over Fig. 4. 1H NMR spectrum of a total lipid extract from an obese SHHF heart rat. Main peaks or regions are assigned as follows: 1, Cholesterol (Chol, C18); 2, CH3 terminal of fatty acyl chain (FA) and Chol (C26, C27); 3, characteristic triplet of CH3 terminal of (n-3) polyunsaturated FA; 4, Chol (C19); 5, (CH2)n of FA; 6, –CH2– CH2–COO–; 7, –CH2–CHfCH–CH2–CHfCH–CH2–; 8, –CH2–COO–; 9, –CHfCH–CH2–CHfCH–; 10, –N+(CH3)3 of phosphatidylcholine; 11, –CH2N+ of phosphatidylcholine; 12, mainly glycerol (C1 and C3) and –O–CH2–CH2N+ of phosphatidylcholine; 13, esterified glycerol (C2); 14, –CHfCH– of FA and Chol (C6); S, solvents (methanol + H2O).
  • 11. 536 J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 Table 6 Quantitation of lipids by NMR SHHF 4 months Lean Acyl chains (nmol/mg w.w.) (n-3) Acyl chains % Cholesterol (nmol/mg w.w.) Choline phospholipids (nmol/mg w.w.) Mean unsaturation Mean poly-unsaturation 103.49 ± 11.28 8.80 ± 0.56 5.09 ± 0.33 14.54 ± 1.00 1.70 ± 0.09 1.17 ± 0.06 SHHF 10 months Obese 117.49 ± 16.87 10.35 ± 0.79 b 5.33 ± 0.95 15.72 ± 2.06 a 1.78 ± 0.12 1.23 ± 0.08 a Lean a Obese 90.74 ± 6.11 8.11 ± 1.25 4.61 ± 0.62 13.57 ± 0.89 1.78 ± 0.10 1.23 ± 0.07 84.49 ± 4.73 9.52 ± 0.62 b 5.11 ± 0.78 12.72 ± 082 1.90 ± 0.02 b 1.33 ± 0.02 b Data are means ± S.E. from 5 hearts in each group. a Obese 4 months is significantly different from obese 10 months, or lean 4-month-old is significantly different from lean 10-month-old, Student's t test p < 0.05. b Obese is significantly different from lean of the same age, Student's t test p < 0.05. background. According to previously published work [30], this lowers the false positive rate to 0.7%. According to these criteria, we did not have any evidence of an effect of obesity in 4- or 10-month-old animals. However, macroarrays analysis on a set of known cardiac genes recapitulated the differential expression of genes involved in hypertension and the development of heart failure (Table 3). Hierarchical clustering analysis of gene expression from lean animals revealed in the same main cluster 3 genes out of 11 encoding proteasome proteins COP9 subunit 4 (CSN4), aminolevulinate deltadehydratase (ALAD) and CHMP1.5 (Table 4A) PKD1 is a cation channel regulator [32], a complex involved in many regulatory processes, including control of development and regulation of morphogenesis. Two genes, UDPGal:betaGlcNAc beta 1,4-galactosyltransferase (B4GALT1) and trichohyalin (THH), encode for proteins that have a structural role. UDP N-acetylglucosamine β-1,4 galactosyltransferase is a widely distributed enzyme which catalyzes the transfer of galactose to N-acetylglucosamine residues of glycoproteins and glycolipids [33]. Elevated B4GALT1 expression was already shown to occur in the failing hearts of spontaneously hypertensive rats [34] and was found down regulated in our experiments that monitored rat heart gene expression prior to HF. In this cluster of down regulated genes, we also observed a member of a novel Ca2+-binding protein subfamily (CABP1), that is a component of Ca2+-mediated cellular signal transduction in the heart [35]. We also noticed a down regulation of genes involved in cell proliferation and/or protein synthesis in muscle and heart tissue such as the RNA binding protein (SEB4D) that was shown to be also down regulated in colon cancer [36]. FGFR1OP, which protects cells from apoptosis, was down regulated as well [37]. Cluster 1 in the obese analysis contained a set of genes encoding remodeling and structural proteins and a second group of genes with designated miscellaneous functions (Table 4B). In the group of genes encoding remodeling and structural proteins, we noticed upregulation of complement factor 1 (CF1). It is well known that both free radicals and complement activation can injure tissue. Local complement activation may represent a mechanism by which free radicals mediate tissue injury [38]. It has been shown that complement activation is directly involved in chronically sustained myocardial damage [39]. Up-regulation of CF1 probably contributes to cardiac remodeling. The second gene is B4GALT1, mentioned above and the third gene is SPOCK2. SPOCK2 encodes a protein called testican-2, which is able to abrogate inhibition of membrane type metalloproteinases [40]. KS1 belongs to the largest family of zinc-finger transcription factors containing the KRAB domain. The functions proposed for members of the KRAB-containing protein family are transcriptional repression of RNA polymerase and binding and splicing of RNA. KS1 counteracts neoplastic transformation induced by several oncogenes [41]. Therefore KS1 could be involved in maintenance of the nucleolus, cell differentiation, cell proliferation, apoptosis and neoplastic transformation as found for other KRAB-containing proteins [42]. In addition, we found heart-type fatty acid binding protein (FABP3) down-regulated in this cluster. FABP3 is involved in lipid metabolism and constitutes a biochemical marker of myocyte injury in HF [43] whose expression was found to be regulated in the heart in response to fatty acid levels [44]. Interestingly, troponin (TNNI3), a well known marker of HF [43], was also found down regulated in this cluster. In the group of miscellaneous genes, we found down regulation of 5′-nucleotidase (NT5). NT5 degrades the adenosine moiety of ATP. Ischemic preconditioning was found to activate NT5. Moreover, it was found that plasma adenosine levels are increased in patients with chronic HF. NT5 activity also increased in the blood and the myocardium in patients with chronic HF, which may explain the increases in adenosine levels in the plasma and the myocardium. In addition, it was found that further elevation of plasma adenosine levels due to either dipyridamole or dilazep reduces the severity of chronic HF. Thus, endogenous adenosine was proposed for cardioprotection in chronic HF and against ischemia and reperfusion injury [45]. The last gene in this cluster is KIR7.1, encoding for an inwardly rectifying potassium (Kir) channel. Kir channels are ubiquitously expressed and serve functions as diverse as regulation of resting membrane potential, maintenance of K+ homeostasis, control of heart rate and hormone secretion [46]. NMR analysis allowed the quantitation of a number of metabolites. Metabolite contents determined were in accordance with published data [47–49]. We noticed a general drop in metabolite concentrations linked to the age of the animals. In addition, the glutamine to glutamate ratio was significantly lowered in obese animals. Globally lowered metabolite levels can be explained by an increase of non-cellular mass such as
  • 12. J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 excessive collagen type I and III that accumulate in the myocardium in hypertensive heart tissue [50]. A lowered glutamine to glutamate ratio could be a consequence of increased TGF-β stimulation since the TGF-β upregulates the phosphate dependant glutaminase and the Na+/H+ exchanger 3 gene expression pathway [51,52] favoring intracellular glutamine conversion to glutamate. Moreover, plasmatic TGF-β circulating levels are increased in obesity [53] and favor extracellular matrix expansion [54]. In addition, this lowered glutamine to glutamate ratio in obese may reflect the use of glutamine to generate more ATP to fuel the cardiomyocytes and to respond to an increased energy need [52]. Fatty acyl chain concentrations were lower in 10-monthold animals when compared to 4-month-old obese animals. In accordance with this observation, FABP3, involved in lipid uptake, was down regulated in obese 10-month-old animals versus 4-month-old (Table 4B). Besides, this apparent lowering of fatty acyl chain concentrations might also be related to the increase of myocardial mass by extracellular matrix accumulation as mentioned above [55]. In accordance with this hypothesis, we observed a kinetic up-regulation of fribronectin gene expression in the obese group (supplemental Table 2 on-line). Fibronectin is known to accumulate in the interstitial space [55]. Since extracellular matrix accumulation is a known occurrence in the remodelling heart and global metabolite levels were lower in obese animals, (except for taurine), this suggests that the obese heart is more fibrotic than its lean counterpart at the same age. This would explain earlier heart failure development in the obese compared to the lean SHHF rat [14]. Interestingly, n-3 fatty acyl chains percentage was higher in obese animals. This observation was confirmed by an increased total fatty acyl unsaturation in obese animals. Hypertension and obesity are known to induce myocardium remodelling [56,57]. Such omega-3 accumulation has already been recently observed during muscle regeneration [27] and may reflect mechanisms involved in the increase of septal wall thickness as observed in the 10 months obese rats (Table 1). The increase of unsaturated lipids was accompanied by conservation of taurine levels (Table 5). Taurine was shown to preserve unsaturated membrane lipids from lipid peroxidation [58]. We propose that cardiac remodelling may share some similarities with muscle regeneration. This increase of omega-3 may contribute to the stimulation of membrane expansion from cardiac cells as observed in PC12 cells [59]. In this present work we could identify a transcriptome kinetic adaptation that was different in lean and obese animals (Figs. 1 and 2) but the 10-month-old lean and obese animals had a very similar transcriptome. In our previous work performed on human right appendage biopsies, we had shown that obese hypertensive patients had clearly distinct cardiac gene expression patterns when compared to hypertensive patients. Thus, several hypotheses are plausible. We could surmise that extreme hypertension in the rat masks most obesity's contribution to gene regulation. In addition, the duration of obesity is much longer in patients than in rats and this point may be important to allow for observation of the contribution of obesity to gene 537 regulations at the transcriptome level in hypertensive animals. Although, leptin is one element among a number of adipokines that are present in obese's bloodstream and that can regulate heart gene expression, the lack of leptin signaling in the SHHF obese rats may have some consequences on specific obesityinduced gene expression pattern. Indeed, it has been observed that leptin added to cardiomyocytes in cell culture can induce hypertrophy [60] and hyperplasia [61]. In hypertensive, nonobese humans, plasmatic levels of leptin have been associated with myocardial wall thickness [62]. However, protein synthesis regulation is known to occur not only at the mRNA level and we did not perform a proteome analysis in this work to investigate for post-transcriptional or post-traductional gene regulations but rather a 1H NMR metabolomic analysis which may be considered as a more downstream observation that takes into account not only the protein level but also its enzymatic activities. 1H NMR metabolomic analysis revealed significant differences in left ventricle metabolites concentration between obese and lean animals. Therefore, this work provides some new insights both at the heart metabolome and transcriptome level during the early steps of heart failure development in the SHHF rat. Acknowledgments We thank Dr Sergueï Sokol (Toulouse Genomic Core Facilities) respectively for array data WEB management. We are indebted to Dr Peter J. Romanienko (Memorial SloanKettering Cancer Center, New York, New York, USA) for critical reading of the manuscript. Appendix A. Supplementary data Supplementary data associated with this article can be found, in the online version, at doi:10.1016/j.yjmcc.2006.11.007. References [1] Basdevant A. Obesity: epidemiology and public health. Ann Endocrinol (Paris) 2000;61(Suppl 6):6–11. [2] Contaldo F, Pasanisi F, Finelli C, de Simone G. Obesity, heart failure and sudden death. Nutr Metab Cardiovasc Dis 2002;12:190–7. [3] Mykkanen L, Kuusisto J, Pyorala K, Laakso M. Cardiovascular disease risk factors as predictors of type 2 (non-insulin-dependent) diabetes mellitus in elderly subjects. Diabetologia 1993;36:553–9. [4] Hubert HB, Feinleib M, McNamara PM, Castelli WP. Obesity as an independent risk factor for cardiovascular disease: a 26-year follow-up of participants in the Framingham Heart Study. Circulation 1983;67: 968–77. [5] Kenchaiah S, Evans JC, Levy D, Wilson PW, Benjamin EJ, Larson MG, Kannel WB, Vasan RS. Obesity and the risk of heart failure. N Engl J Med 2002;347:305–13. [6] Kagaya Y, Kanno Y, Takeyama D, Ishide N, Maruyama Y, Takahashi T, Ido T, Takishima T. Effects of long-term pressure overload on regional myocardial glucose and free fatty acid uptake in rats. A quantitative autoradiographic study. Circulation 1990;81:1353–61. [7] Beske SD, Alvarez GE, Ballard TP, Davy KP. Reduced cardiovagal baroreflex gain in visceral obesity: implications for the metabolic syndrome. Am J Physiol Heart Circ Physiol 2002;282:H630–5. [8] Ahima RS, Flier JS. Adipose tissue as an endocrine organ. Trends Endocrinol Metab 2000;11:327–32.
  • 13. 538 J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 [9] Guerre-Millo M. Adipose tissue and adipokines: for better or worse. Diabetes Metab 2004;30:13–9. [10] Guerre-Millo M. Adipose tissue hormones. J Endocrinol Invest 2002;25:855–61. [11] Philip-Couderc P, Smih F, Pelat M, Vidal C, Verwaerde P, Pathak A, et al. Cardiac transcriptome analysis in obesity-related hypertension. Hypertension 2003;41:414–21. [12] Philip-Couderc P, Smih F, Hall JE, Pathak A, Roncalli J, Harmancey R, et al. Kinetic analysis of cardiac transcriptome regulation during chronic high-fat diet in dogs. Physiol Genomics 2004;19:32–40. [13] Philip-Couderc P, Pathak A, Smih F, Dambrin C, Harmancey R, Buys S, et al. Uncomplicated human obesity is associated with a specific cardiac transcriptome: involvement of the Wnt pathway. FASEB J 2004;18: 1539–40. [14] McCune S, Baker P, Still H. SHHF/Mcc-cp rat: model of obesity, noninsulin-dependent diabetes, and congestive heart failure. ILAR News 1990;32:23–7. [15] Heyen JR, Blasi ER, Nikula K, Rocha R, Daust HA, Frierdich G, et al. Structural, functional, and molecular characterization of the SHHF model of heart failure. Am J Physiol Heart Circ Physiol 2002;283:H1775–84. [16] Wheeler DL, Church DM, Federhen S, Lash AE, Madden TL, Pontius JU, et al. Database resources of the National Center for Biotechnology. Nucleic Acids Res 2003;31:28–33. [17] Liang F, Holt I, Pertea G, Karamycheva S, Salzberg SL, Quackenbush J. Gene index analysis of the human genome estimates approximately 120,000 genes. Nat Genet 2000;25:239–40. [18] Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, et al. TM4: a free, open-source system for microarray data management and analysis. BioTechniques 2003;34:374–8. [19] Rajeevan MS, Ranamukhaarachchi DG, Vernon SD, Unger ER. Use of real-time quantitative PCR to validate the results of cDNA array and differential display PCR technologies. Methods 2001;25:443–51. [20] Schmittgen TD, Zakrajsek BA. Effect of experimental treatment on housekeeping gene expression: validation by real-time, quantitative RTPCR. J Biochem Biophys Methods 2000;46:69–81. [21] Aerts JL, Gonzales MI, Topalian SL. Selection of appropriate control genes to assess expression of tumor antigens using real-time RT-PCR. BioTechniques 36 2004; 84–86, 88, 90–91. [22] Tyagi RK, Azrad A, Degani H, Salomon Y. Simultaneous extraction of cellular lipids and water-soluble metabolites: evaluation by NMR spectroscopy. Magn Reson Med 1996;35:194–200. [23] Desmoulin F, Confort-Gouny S, Masson S, Bernard M, Doddrell DM, Cozzone PJ. Application of reverse-DEPT polarization transfer pulse sequence to study the metabolism of carbon-13-labeled substrates in perfused organs by 1H NMR spectroscopy. Magn Reson Med 1990;15: 456–61. [24] Bollard ME, Murray AJ, Clarke K, Nicholson JK, Griffin JL. A study of metabolic compartmentation in the rat heart and cardiac mitochondria using high-resolution magic angle spinning 1H NMR spectroscopy. FEBS Lett 2003;553:73–8. [25] Mayr M, Metzler B, Chung YL, McGregor E, Mayr U, Troy H, et al. Ischemic preconditioning exaggerates cardiac damage in PKC-delta null mice. Am J Physiol Heart Circ Physiol 2004;287:H946–56. [26] Adosraku RK, Choi GT, Constantinou-Kokotos V, Anderson MM, Gibbons WA. NMR lipid profiles of cells, tissues, and body fluids: proton NMR analysis of human erythrocyte lipids. J Lipid Res 1994;35:1925–31. [27] Gillet B, Sebrie C, Bogaert A, Bleneau S, de la Porte S, Beloeil JC. Study of muscle regeneration using in vitro 2D 1H spectroscopy. Biochim Biophys Acta 2005;1724:333–44. [28] Astrakas LG, Goljer I, Yasuhara S, Padfield KE, Zhang Q, Gopalan S, et al. Proton NMR spectroscopy shows lipids accumulate in skeletal muscle in response to burn trauma-induced apoptosis. FASEB J 2005;19:1431–40. [29] Siddiqui N, Sim J, Silwood CJ, Toms H, Iles RA, Grootveld M. Multicomponent analysis of encapsulated marine oil supplements using high-resolution 1H and 13C NMR techniques. J Lipid Res 2003;44: 2406–27. [30] Liu T, Lai H, Wu W, Chinn S, Wang PH. Developing a strategy to define [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] the effects of insulin-like growth factor-1 on gene expression profile in cardiomyocytes. Circ Res 2001;88:1231–8. Haas GJ, McCune SA, Brown DM, Cody RJ. Echocardiographic characterization of left ventricular adaptation in a genetically determined heart failure rat model. Am Heart J 1995;130:806–11. Vandorpe DH, Chernova MN, Jiang L, Sellin LK, Wilhelm S, Stuart-Tilley AK, et al. The cytoplasmic C-terminal fragment of polycystin-1 regulates a Ca2+-permeable cation channel. J Biol Chem 2001;276:4093–101. Shaper NL, Mann PL, Shaper JH. Cell surface galactosyltransferase: immunochemical localization. J Cell Biochem 1985;28:229–39. Humphries DE, Sirokman G, Bing OH. Enhanced galactosyltransferase expression in the failing hearts of spontaneously hypertensive rats. Biochem Biophys Res Commun 1996;218:320–4. Haeseleer F, Sokal I, Verlinde CL, Erdjument-Bromage H, Tempst P, Pronin AN, et al. Five members of a novel Ca(2+)-binding protein (CABP) subfamily with similarity to calmodulin. J Biol Chem 2000;275:1247–60. Scanlan MJ, Welt S, Gordon CM, Chen YT, Gure AO, Stockert E, et al. Cancer-related serological recognition of human colon cancer: identification of potential diagnostic and immunotherapeutic targets. Cancer Res 2002;62:4041–7. Guasch G, Ollendorff V, Borg JP, Birnbaum D, Pebusque MJ. 8p12 stem cell myeloproliferative disorder: the FOP-fibroblast growth factor receptor 1 fusion protein of the t(6;8) translocation induces cell survival mediated by mitogen-activated protein kinase and phosphatidylinositol 3-kinase/ Akt/mTOR pathways. Mol Cell Biol 2001;21:8129–42. Tanhehco EJ, Yasojima K, McGeer PL, Washington RA, Lucchesi BR. Free radicals upregulate complement expression in rabbit isolated heart. Am J Physiol Heart Circ Physiol 2000;279:H195–201. Yasojima K, Schwab C, McGeer EG, McGeer PL. Human heart generates complement proteins that are upregulated and activated after myocardial infarction. Circ Res 1998;83:860–9. Nakada M, Miyamori H, Yamashita J, Sato H. Testican 2 abrogates inhibition of membrane-type matrix metalloproteinases by other testican family proteins. Cancer Res 2003;63:3364–9. Gebelein B, Fernandez-Zapico M, Imoto M, Urrutia R. KRAB-independent suppression of neoplastic cell growth by the novel zinc finger transcription factor KS1. J Clin Invest 1998;102:1911–9. Urrutia R. KRAB-containing zinc-finger repressor proteins. Genome Biol 2003;4:231. Sato Y, Kita T, Takatsu Y, Kimura T. Biochemical markers of myocyte injury in heart failure. Heart 2004;90:1110–3. Van Bilsen M, de Vries JE, Van der Vusse GJ. Long-term effects of fatty acids on cell viability and gene expression of neonatal cardiac myocytes. Prostaglandins Leukot Essent Fatty Acids 1997;57:39–45. Kitakaze M, Minamino T, Node K, Takashima S, Funaya H, Kuzuya T, et al. Adenosine and cardioprotection in the diseased heart. Jpn Circ J 1999;63:231–43. Abraham MR, Jahangir A, Alekseev AE, Terzic A. Channelopathies of inwardly rectifying potassium channels. FASEB J 1999;13:1901–10. Decker R, von Stuckrad-Barre S, Milakofsky L, Hofford JM, Harris N, Vogel WH. Effect of stress on amino acids and related compounds in various tissues of the rat. Life Sci 1995;57:1781–90. Nielsen LB, Perko M, Arendrup H, Andersen CB. Microsomal triglyceride transfer protein gene expression and triglyceride accumulation in hypoxic human hearts. Arterioscler Thromb Vasc Biol 2002;22:1489–94. Michael O'Donnell J, Narayan P, Bailey MQ, Abduljalil AM, Altschuld RA, McCune SA, et al. 31P-NMR analysis of congestive heart failure in the SHHF/Mcc-facp rat heart. J Mol Cell Cardiol 1998;30:235–41. Diez J, Gonzalez A, Lopez B, Querejeta R. Mechanisms of disease: pathologic structural remodeling is more than adaptive hypertrophy in hypertensive heart disease. Nat Clin Pract Cardiovasc Med 2005;2: 209–16. Nelson D, Rumsey WL, Erecinska M. Glutamine catabolism by heart muscle. Properties of phosphate-activated glutaminase. Biochem J 1992 (Mar 1);282(Pt 2):559–64. Welbourne T, Routh R, Yudkoff M, Nissim I. The glutamine/glutamate couplet and cellular function. News Physiol Sci 2001;16:157–60. Peterson MC. Circulating transforming growth factor beta-1: a partial
  • 14. J. Roncalli et al. / Journal of Molecular and Cellular Cardiology 42 (2007) 526–539 [54] [55] [56] [57] [58] molecular explanation for associations between hypertension, diabetes, obesity, smoking and human disease involving fibrosis. Med Sci Monit 2005;11:RA229–32. Lijnen PJ, Petrov VV, Fagard RH. Induction of cardiac fibrosis by transforming growth factor-beta(1). Mol Genet Metab 2000;71:418–35. Sharov VG, Kostin S, Todor A, Schaper J, Sabbah HN. Expression of cytoskeletal, linkage and extracellular proteins in failing dog myocardium. Heart Fail Rev 2005;10:297–303. Gosse P, Dallocchio M. Left ventricular hypertrophy: epidemiological prognosis and associated critical factors. Eur Heart J 1993;14(Suppl D):16–21. Wakatsuki T, Schlessinger J, Elson EL. The biochemical response of the heart to hypertension and exercise. Trends Biochem Sci 2004;29: 609–17. Nandhini AT, Thirunavukkarasu V, Ravichandran MK, Anuradha CV. [59] [60] [61] [62] 539 Effect of taurine on biomarkers of oxidative stress in tissues of fructose-fed insulin-resistant rats. Singapore Med J 2005;46:82–7. Darios F, Davletov B. Omega-3 and omega-6 fatty acids stimulate cell membrane expansion by acting on syntaxin 3. Nature 2006;440: 813–7. Rajapurohitam V, Gan XT, Kirshenbaum LA, Karmazyn M. The obesityassociated peptide leptin induces hypertrophy in neonatal rat ventricular myocytes. Circ Res 2003;93:277–9. Tajmir P, Ceddia RB, Li RK, Coe IR, Sweeney G. Leptin increases cardiomyocyte hyperplasia via extracellular signal-regulated kinase- and phosphatidylinositol 3-kinase-dependent signaling pathways. Endocrinology 2004;145:1550–5. Paolisso G, Tagliamonte MR, Galderisi M, Zito GA, Petrocelli A, Carella C, et al. Plasma leptin level is associated with myocardial wall thickness in hypertensive insulin-resistant men. Hypertension 1999;34:1047–52.