ArticlePDF Available

The Novel Mas agonist, CGEN-856S, Attenuates Isoproterenol-Induced Cardiac Remodeling and Myocardial Infarction Injury in Rats

PLOS
PLOS ONE
Authors:

Abstract and Figures

CGEN-856S is a novel Mas agonist. Herein, we examined the effects of this peptide on isoproterenol (ISO)-induced cardiac remodeling and myocardial infarction (MI) injury. We also sought to determine whether CGEN-856S activates the underlying mechanisms related to Mas receptor activation. Heart hypertrophy and fibrosis were induced by ISO (2 mg·kg·day) in Wistar rats. After a 7-day treatment period with CGEN-856S (90 µg·kg·day) or vehicle, the cardiomyocyte diameter was evaluated in left ventricular sections stained with hematoxylin and eosin, and immunofluorescence labeling and quantitative confocal microscopy were used to quantify the deposition of type I and III collagen and fibronectin in the left ventricles. MI was induced by coronary artery ligation, and CGEN-856S (90 µg·kg·day) or saline was administered for 14 days. The Langendorff technique was used to evaluate cardiac function, and left ventricular sections were stained with Masson's trichrome dye to quantify the infarct area. Using Chinese hamster ovary cells stably transfected with cDNA, we evaluated whether CGEN-856S alters AKT and endothelial nitric oxide synthase (eNOS) phosphorylation. CGEN-856S reduced the degree of ISO-induced hypertrophy (13.91±0.17 µm vs. 12.41±0.16 µm in the ISO+CGEN-856S group). In addition, the Mas agonist attenuated the ISO-induced increase in collagen I, collagen III, and fibronectin deposition. CGEN-856S markedly attenuated the MI-induced decrease in systolic tension, as well as in +dT/dt and -dT/dt. Furthermore, CGEN-856S administration significantly decreased the infarct area (23.68±2.78% vs. 13.95±4.37% in the MI+CGEN-856S group). These effects likely involved the participation of AKT and NO, as CGEN-856S administration increased the levels of p-AKT and p-eNOS. Thus, our results indicate that CGEN-856S exerts cardioprotective effects on ISO-induced cardiac remodeling and MI-mediated heart failure in rats through a mechanism likely involving the eNOS/AKT pathway.
Content may be subject to copyright.
The Novel Mas agonist, CGEN-856S, Attenuates
Isoproterenol-Induced Cardiac Remodeling and
Myocardial Infarction Injury in Rats
´lvia Q. Savergnini
1
, Danielle Ianzer
1
, Mariana B. L. Carvalho
1
, Anderson J. Ferreira
2
, Gerluza A. B. Silva
2
,
Fu
´lvia D. Marques
1
, Anto
ˆnio Augusto B. Peluso
1
, Merav Beiman
3
, Gady Cojocaru
3
, Yossi Cohen
3
,
Alvair P. Almeida
1
, Galit Rotman
3
, Robson A. S. Santos
1
*
1Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil, 2Department of Morphology, Federal University of Minas
Gerais, Belo Horizonte, MG, Brazil, 3Compugen Ltd., Tel Aviv, Israel
Abstract
CGEN-856S is a novel Mas agonist. Herein, we examined the effects of this peptide on isoproterenol (ISO)-induced cardiac
remodeling and myocardial infarction (MI) injury. We also sought to determine whether CGEN-856S activates the underlying
mechanisms related to Mas receptor activation. Heart hypertrophy and fibrosis were induced by ISO (2 mg?kg
21
?day
21
)in
Wistar rats. After a 7-day treatment period with CGEN-856S (90 mg?kg
21
?day
21
) or vehicle, the cardiomyocyte diameter was
evaluated in left ventricular sections stained with hematoxylin and eosin, and immunofluorescence labeling and
quantitative confocal microscopy were used to quantify the deposition of type I and III collagen and fibronectin in the left
ventricles. MI was induced by coronary artery ligation, and CGEN-856S (90 mg?kg
21
?day
21
) or saline was administered for 14
days. The Langendorff technique was used to evaluate cardiac function, and left ventricular sections were stained with
Masson’s trichrome dye to quantify the infarct area. Using Chinese hamster ovary cells stably transfected with Mas cDNA, we
evaluated whether CGEN-856S alters AKT and endothelial nitric oxide synthase (eNOS) phosphorylation. CGEN-856S
reduced the degree of ISO-induced hypertrophy (13.9160.17 mm vs. 12.4160.16 mm in the ISO+CGEN-856S group). In
addition, the Mas agonist attenuated the ISO-induced increase in collagen I, collagen III, and fibronectin deposition. CGEN-
856S markedly attenuated the MI-induced decrease in systolic tension, as well as in +dT/dt and -dT/dt. Furthermore, CGEN-
856S administration significantly decreased the infarct area (23.6862.78% vs. 13.9564.37% in the MI+CGEN-856S group).
These effects likely involved the participation of AKT and NO, as CGEN-856S administration increased the levels of p-AKT and
p-eNOS. Thus, our results indicate that CGEN-856S exerts cardioprotective effects on ISO-induced cardiac remodeling and
MI-mediated heart failure in rats through a mechanism likely involving the eNOS/AKT pathway.
Citation: Savergnini SQ, Ianzer D, Carvalho MBL, Ferreira AJ, Silva GAB, et al. (2013) The Novel Mas agonist, CGEN-856S, Attenuates Isoproterenol-Induced Cardiac
Remodeling and Myocardial Infarction Injury in Rats. PLoS ONE 8(3): e57757. doi:10.1371/journal.pone.0057757
Editor: Michael Bader, Max-Delbru
¨ck Center for Molecular Medicine (MDC), Germany
Received October 29, 2012; Accepted January 24, 2013; Published March 1, 2013
Copyright: ß2013 Savergnini et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: S.Q.S. was supported by a fellowship from Conselho Nacional de Desenvolvimento Cientı
´fico e Tecnolo
´gico (CNPq) and F.D.M. was supported by
a fellowship from Coordenac¸a
˜o de Aperfeic¸oamento de Pessoal de Nı
´vel Superior (CAPES). This study was supported by a Compugen Grant and by Ministe
´rio de
Cie
ˆncia e Tecnologia/Fundac¸a
˜o de Amparo a
`Pesquisa do Estado de Minas Gerais (Fapemig)/Instituto Nacional de Cie
ˆncia e Tecnologia-INCT-NanoBiofar. The
funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors would like to declare that Merav Beiman, Gady Cojocaru, Yossi Cohen and Galit Rotman are affiliated to the commercial
funders of this research (Compugen Ltd.) and this does not alter the authors’ adherence to all the PLOS ONE policies on sharing data and materials.
* E-mail: robsonsant@gmail.com
Introduction
Cardiac remodeling is an adaptive response to the pathogenesis
of several heart diseases that interferes with the function and
structure of the myocardium [1–3]. This structural remodeling
process predisposes patients to an increased risk of adverse cardiac
events, including myocardial ischemia, myocardial infarction (MI),
arrhythmias, and sudden cardiac death [3]. A growing body of
evidence indicates that the renin-angiotensin system (RAS) plays
an important role in the development and progression of cardiac
remodeling. Angiotensin (Ang) II, the main end product of the
RAS cascade, stimulates the biosynthesis of cardiac extracellular
matrix (ECM) proteins, leading to interstitial and perivascular
fibrosis [4,5], and cardiomyocyte hypertrophy [6]. On the other
hand, numerous studies have proposed that the cardioprotective
axis of the RAS composed by Ang-converting enzyme (ACE) 2,
Ang-(1–7), and the Mas receptor counterregulates these Ang II
actions in the heart [7–11]. Indeed, it has been reported that Ang-
(1–7) reduces the growth of cardiac myocytes through activation of
the G protein-coupled receptor (GPCR) Mas [12] and inhibits
cardiac fibroblast-mediated collagen deposition [13]. Additionally,
the hearts of Mas-deficient mice exhibit marked changes in ECM
protein expression leading to a profibrotic profile accompanied by
cardiac dysfunction [14,15]. Grobe et al. [8,16] demonstrated that
Ang-(1–7) prevented cardiac fibrosis elicited by either deoxycorti-
costerone acetate (DOCA)-salt treatment or Ang II infusion,
independent of blood pressure changes. Moreover, AVE 0991,
a nonpeptide Ang-(1–7) analog, prevents the development of
isoproterenol (ISO)-induced hypertrophy and collagen deposition
[17]. Recently, He et al. [18] reported that AVE 0991 prevents
Ang II-induced myocardial hypertrophy in a dose-dependent
manner. Taken together, these findings indicate that the ACE2/
PLOS ONE | www.plosone.org 1 March 2013 | Volume 8 | Issue 3 | e57757
Ang-(1–7)/Mas axis is involved in the prevention and attenuation
of cardiac remodeling.
The evidence supporting the cardioprotective effects of the
ACE2/Ang-(1–7)/Mas axis prompted us to search for novel Mas
ligands. We used a computational biology discovery platform that
utilizes machine learning algorithms designed to predict novel
GPCR ligands cleaved from secreted proteins by convertase
proteolysis, which are extracted from the Swiss-Prot protein
database [19,20]. The predicted ligands were synthesized and
screened for activation of 152 GPCRs using calcium flux and
cyclic adenosine monophosphate (cAMP) assays [20]. Two novel
peptide ligands, P61S and P33V, displayed high specificity for
Mas, eliciting calcium influx in Mas-overexpressing Chinese
hamster ovary (CHO) cells [20]. Furthermore, P61S, designated
CGEN-856S, does not activate either Ang II type 1 (AT
1
) or type
2 (AT
2
) receptors [20,21].
We recently reported that CGEN-856S elicits nitric oxide (NO)-
dependent vasodilation mediated by Mas in rat and mice aorta
rings [21]. Additionally, picomolar concentrations of this peptide
(40 pmol/L) induced an antiarrhythmogenic effect, as demon-
strated by reduced incidence and duration of reperfusion
arrhythmias in isolated rat hearts [21]. In addition, acute
CGEN-856S administration produced a dose-dependent decrease
in the blood pressure of spontaneously hypertensive rats (SHR)
[21]. Importantly, the actions of CGEN-856S are inhibited by the
Mas antagonist A-779 or by genetic deletion of this receptor [21].
In general, the data obtained following the use of this peptide
resemble to those reported previously for Ang-(1–7) and AVE
0991. Thus, in the present study, we hypothesized that CGEN-
856S might mimic the antihypertrophic and antifibrotic effects
induced by Ang-(1–7) in rat hearts. To test this hypothesis, we
evaluated the cardiac structure of ISO-treated and of infarcted
rats. In addition, the effects of CGEN-856S administration on
AKT and endothelial NO synthase (e-NOS) activation were
investigated.
Materials and Methods
Ethics Statement and Animals
Male Wistar rats weighing 2402300 g were obtained from the
animal facility of the Biological Sciences Institute of the Federal
University of Minas Gerais. The experimental protocols were
approved by the Ethics Committee in Animal Experimentation of
the Federal University of Minas Gerais, Brazil (CETEA-UFMG),
in accordance with the National Institutes of Health (NIH)
Guidelines for the Care and Use of Laboratory Animals (protocol
#149/10).
Isoproterenol Induction of Hypertrophy
Osmotic mini-pumps (AlzetH, model 2001) containing CGEN-
856S (90 mg?kg
21
?day
21
, 7 days) or saline were implanted
subcutaneously (sc) under anesthesia (10% ketamine/2% xylazine,
0.1 mL/100 g, intraperitoneally [ip]). After recovery from anes-
thesia, the animals were divided into 4 groups: i) saline +vehicle
(olive oil, 1 mL?g
21
?day
21
, sc, 7 days), ii) saline +ISO
(2 mg?kg
21
?day
21
, sc, 7 days), iii) CGEN-856S +vehicle; and
(iv) CGEN-856S +ISO. Moreover, as a positive control, an
additional group of rats were treated with losartan
(1 mg?kg
21
?day
21
, gavage, 7 days) +vehicle (olive oil,
1mL?kg
21
?day
21
, sc, 7 days) or losartan (1 mg?kg
21
?day
21
,
gavage, 7 days) +ISO (2 mg?kg
21
?day
21
, sc, 7 days). The final
gavage and sc injection volumes were approximately 0.5 and
0.2 mL, respectively.
Histological Analysis
At the end of the 7-day ISO treatment period, the rats were
sacrificed by decapitation and the hearts were immediately
removed. The left ventricles were fixed in 4% paraformaldehyde
for 48 h at room temperature. The tissues were dehydrated by
sequential washes with 70%, 80%, 90%, and 100% ethanol and
embedded in ParaplastHX-tra Tissue Embedding Medium
(McCormick Scientific). Transversal sections (6 mm) were cut
starting from the base area of the left ventricle at 40-mm intervals
and stained with hematoxylin and eosin for cell morphometry
(n = 4 for each group). The cardiomyocyte diameter was evaluated
in the tissue sections (324 for each animal) using an ocular
micrometer calibrated with a stage micrometer adapted to a light
microscope (BX 60, Olympus) at 1006magnification and analyzed
using Image Pro Express software. Only cardiomyocytes cut
longitudinally with the nuclei and cellular limits visible were used
for analysis (an average of 15 cardiomyocytes for each slice). The
diameter of each myocyte was measured across the region
corresponding to the nucleus. Approximately 50 cardiomyocytes
were analyzed for each animal.
Immunofluorescence Analysis
Immunofluorescence labeling and quantitative confocal micros-
copy were used to investigate the distribution and quantity of type
I and III collagen and fibronectin present in the left ventricles of
the animals included in the ISO protocol (n = 425 rats/group).
The hearts were enclosed in Tissue Tek OCT compound (Miles
Scientific, Chicago, IL, USA), immediately frozen in liquid
nitrogen, and stored at -80uC. Ventricular sections (7 mm) were
obtained using a cryostat at -20uC, mounted on slides, fixed with
ethanol for 10 min, and dried at room temperature. The slides
were rehydrated with phosphate-buffered saline (PBS) for 10 min
and incubated in blocking solution (1% bovine serum albumin
[BSA] and 0.1% Tween 20 in PBS) at room temperature for
30 min. The sections were incubated overnight at 4uC with one of
the following primary antibodies: rabbit anti-human type I
collagen (1:100, Rockland), rabbit anti-human type III collagen
(1:100, Rockland), or rabbit anti-human fibronectin (1:200,
Rockland). All antibodies were diluted with a 1:10 dilution of
blocking solution. After 425 PBS rinses, donkey anti-rabbit
immunoglobulin G (IgG) conjugated with Alexa Fluor 488
Figure 1. Effects of CGEN-856S and losartan administration on
the cardiomyocyte diameters of isoproterenol-treated rats.
Animals were treated with isoproterenol (ISO) for 7 days to induce heart
hypertrophy or with olive oil as a control. The effects of CGEN-856S
were compared to those of saline as a negative control (Veh) or losartan
(LOS) as a positive control. Values are expressed as mean 6standard
error of the mean (SEM), n = 425 animals. *P,0.05 vs. oil +Veh;
#
P,0.05 vs. ISO+Veh;
a
P,0.05 vs. ISO+LOS.
doi:10.1371/journal.pone.0057757.g001
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 2 March 2013 | Volume 8 | Issue 3 | e57757
Figure 2. Effects of CGEN-856S and losartan administration on the deposition of type I collagen (CO I), type III collagen (CO III), and
fibronectin (FN) in the left ventricles of isoproterenol (ISO)-treated rats. (A) Representative confocal photomicrographs and (B)
quantification of CO I, CO III, and FN in the left ventricles of animals treated with CGEN-856S. (C) Representative confocal photomicrographs and (D)
quantification of CO I, CO III, and FN in the left ventricles of animals treated with losartan. Values are expressed as arbitrary units (mean gray value 6
SEM, n = 425 animals). *P,0.05 vs. ISO+vehicle.
doi:10.1371/journal.pone.0057757.g002
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 3 March 2013 | Volume 8 | Issue 3 | e57757
(1:200, Molecular Probes) and DRAQ5 (1:1000, Biostatus) were
added for 1 h in the dark at room temperature. Following PBS
washes, the sections were mounted in 25% glycerol/75% PBS and
viewed with a laser scanning confocal microscope (Zeiss 510
Meta). Optimal confocal settings (aperture, gain, and laser power)
were determined at the beginning of each imaging session and
Figure 3. Effects of CGEN-856S and captopril administration on (A) systolic tension, (B) diastolic tension, (C) +dT/dt, (D) -dT/dt, (E)
coronary flow, and (F) heart rate of rat hearts with myocardial infarction (MI). Values are expressed as mean 6SEM, n = 728 animals.
*P,0.05 vs. sham;
#
P,0.05 vs. MI+vehicle;
a
P,0.05 vs. MI+captopril.
doi:10.1371/journal.pone.0057757.g003
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 4 March 2013 | Volume 8 | Issue 3 | e57757
then held constant during the analysis of all samples. For
quantitative analysis of collagen I and III and fibronectin, we
used ImageTool 2.0 image analysis software to measure the
fluorescence intensity of the randomly selected images. The 12-bit
images were captured and analyzed in the gray scale range of
02255. The fluorescence intensity was calculated as an average of
the area (i.e., the sum of the gray values of all pixels divided by the
number of pixels in the area) and the values were recorded as
arbitrary units.
Myocardial Infarction Procedure
Under anesthesia with 10% ketamine/2% xylazine (0.1 mL/
100 g, ip), the rats were placed in the supine position on a surgical
table, tracheotomized, intubated, and ventilated with room air
using a respirator for small rodents. Subdermal electrodes were
placed for electrocardiography (ECG). The chest was opened by
left thoracotomy at the fourth or fifth intercostal space. To expose
the heart, a small retractor was used to maintain rib separation.
After pericardial incision, the heart was quickly removed from the
Figure 4. Effects of CGEN-856S and captopril administration on left ventricular infarct area. (A) Representative photomicrographs and (B)
quantification of the infarct area of animals treated with CGEN-856S or captopril. Values are expressed as mean 6SEM, n = 728 animals. MI:
myocardial infarction. *P,0.05 vs. sham;
#
P,0.05 vs. MI+vehicle.
doi:10.1371/journal.pone.0057757.g004
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 5 March 2013 | Volume 8 | Issue 3 | e57757
Figure 5. Effects of CGEN-856S and Ang-(1–7) administration on AKT phosphorylation and on the quantity of p-AKT. (A)
Representative immunoblots demonstrating the presence of Mas in Mas-transfected CHO cells (CHO-Mas) and the absence of Mas in untransfected
cells (CHO-K1). (B) Effects of CGEN-856S and Ang-(1–7) administration (10
29
and 10
27
mol/L) for 10 min on p-AKT levels in CHO-Mas cells. (C) The
absence of effects of CGEN-856S and Ang-(1–7) administration (10
27
mol/L) for 10 min on p-AKT levels in CHO-K1 cells. (D) Effects of CGEN-856S and
Ang-(1–7) administration (10
29
mol/L) for 5 min on AKT phosphorylation in CHO-Mas cells. Ang-(1–7) (10
29
and 10
27
mol/L) was used as a positive
control and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and total AKT were used as loading controls. *P,0.05 vs. control. Results are
expressed as the mean 6SEM of 426 experiments.
doi:10.1371/journal.pone.0057757.g005
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 6 March 2013 | Volume 8 | Issue 3 | e57757
Figure 6. Effects of CGEN-856S and Ang-(1–7) administration on p-eNOS levels. CGEN-856S and Ang-(1–7) administration (10
27
mol/L)
increased the quantity of (A) p-eNOS Ser1177, but not (B) p-eNOS Thr495 in CHO-Mas cells. (C) The absence of the effects of CGEN-856S and Ang-(1–
7) administration (10
27
mol/L) on p-eNOS Ser1177 levels in CHO-K1 cells. Cells were exposed to the agonist for 10 min. Ang-(1–7) was used as
a positive control and GAPDH was used as a loading control. *P,0.05 vs. control. Results are expressed as the mean 6SEM of 426 experiments.
doi:10.1371/journal.pone.0057757.g006
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 7 March 2013 | Volume 8 | Issue 3 | e57757
thoracic cavity and turned to the left to allow access to the
proximal left anterior descending (LAD) coronary artery. A 4-0 silk
suture was snared around the LAD and tightly ligated to occlude
the vessel. To increase the survival rate of the animals, coronary
ligation was performed on a more distal portion of the LAD. The
heart was then replaced and the chest was closed with 4-0 silk
sutures. Sham-operated rats were treated in the same manner,
although the coronary artery was not ligated. After the surgical
procedures, ECG tracings were obtained to confirm myocardial
ischemia, i.e., ST-segment elevation and increased R-wave
amplitude. Infarcted rats received CGEN-856S
(90 mmg?kg
21
?day
21
) or vehicle (saline) administered through
osmotic mini-pumps (AlzetH, model 2002) for 14 days. An
additional group of infarcted rats received captopril
(1 mg?kg
21
?day
21
, 14 days) through daily gavage. Fourteen days
after infarction induction, the rats were sacrificed and cardiac
function was evaluated.
Isolated Heart Preparation
The animals (n = 728 rats/group) were decapitated 10215 min
after ip injection of 400 IU of heparin. The thorax was opened,
then the heart was carefully dissected and perfused with Krebs-
Ringer solution (KRS) containing (in mmol/L): 118.4 NaCl,
4.7 KCl, 1.2 KH
2
PO
4
, 1.2 MgSO
4
,7H
2
O, 2.5 CaCl
2
,2H
2
O,
11.7 glucose, and 26.5 NaHCO
3
. The perfusion fluid was
maintained at 3761uC with a pressure of 65275 mmHg and
constant oxygenation (5% CO
2
/95% O
2
). A force transducer was
attached through a heart clip to the apex of the ventricles to record
the contractile force (tension, g) on a computer by a data-
acquisition system (Biopac System). A diastolic tension of
1.060.2 g was applied to the hearts. Electrical activity was
recorded on ECG (Nihon Kohden) with the aid of 2 cotton wicks
placed directly on the surface of the right atrium and left ventricle.
Coronary flow was measured every 5 min by collecting and
determining the volume of heart effluent during a 1-min interval.
After 15220 min of stabilization, the functional parameters
(systolic tension, diastolic tension, 6dT/dt, heart rate, and
coronary flow) were recorded for an additional 30-min period.
Quantification of the Myocardial Infarct Area
At the end of the perfusion, the left ventricles (n =628 for each
group) were fixed in 4% Bouin’s fixative for 24 h at room
temperature. The tissues were dehydrated by sequential washes
with 70%, 80%, and 90% ethanol, 3 washes with 100% ethanol,
and 3 washes with xylene, and then imbedded in paraffin.
Transverse sections (6 mm) of the left ventricles were cut starting
from the median area immediately below the left coronary artery
ligation at 40-mm intervals and stained with Masson’s trichrome to
quantify the infarct area. The infarct area was measured in 2 tissue
sections (both at the median area, one proximal and the other
distal to the coronary ligation of the left ventricle) of each animal.
Images (406magnification) were obtained using a JVC TK-1270/
RGB microcamera. The built-in KS300 software built of
a Kontron Elektronick/Carl Zeiss image analyzer was used for
infarct area quantification using the image segmentation function.
The data were expressed as mm
2
.
Cell Culture and Western Blot Analysis
CHO cells (American Type Culture Collection) stably trans-
fected with Mas and selected by neomycin (CHO-Mas) were
serum-starved 3 h before all experiments. Cells were stimulated for
10 min in serum-free Dulbecco’s modified Eagle’s medium
(DMEM) F-12 medium (Sigma-Aldrich, St. Louis, MO, USA)
with Ang-(1–7) (10
27
mol/L and 10
29
mol/L) or CGEN-856S
(10
27
mol/L and 10
29
mol/L) (Compugen Ltd., Israel). Optimal
conditions, such as the duration of the incubation and peptide
concentration, were chosen based on our initial concentration-
response studies [22]. Untransfected CHO cells were used as
controls and submitted to similar experimental conditions.
After the incubation period, the cells were washed with PBS to
remove metabolic residues and most of the floating cells. The
remaining cells were scraped into 180 mL of lysis buffer (50 mM
Na
4
P
2
O
7
, 50 mM NaF, 5 mM Na
2
EDTA, 5 mM NaCl, 5 mM
EGTA, 10 mM HEPES, 1% Triton X-100, and a specific EDTA-
free inhibitor cocktail) for each cell culture flask (75 cm
2
). The
lysate was transferred to a 1.5 mL tube, homogenized, centrifuged
at 14000 rpm for 20 min at 4uC, and the supernatant was
transferred for another tube. The protein concentration was
assayed using the Bradford protein method. Sixty micrograms of
protein were loaded on a 10% sodium dodecyl sulfate (SDS)
polyacrylamide gel, electrophoresed, and transferred to a nitrocel-
lulose membrane (Bio-Rad, Hercules, CA, USA). The membranes
were blocked in 5% dry milk for 1 h and incubated overnight with
one of the following primary antibodies at 4uC: total AKT (1:1000,
Cell Signaling Technology, Danvers, MA, USA), p-AKT Ser473
(1:500, Cell Signaling Technology, Danvers, MA, USA), p-eNOS
Ser1177 (1:500, Cell Signaling Technology, Danvers, MA, USA),
p-eNOS Thr495 (1:500, Cell Signaling Technology, Danvers,
MA, USA), glyceraldehyde 3-phosphate dehydrogenase (GAPDH)
(1:1000, Cell Signaling Technology, Danvers, MA, USA), and
Mas (1:500) [22]. The secondary antibody was added for 1 h at
room temperature. Protein band detection was performed using
the Odyssey scanning system (Li-Cor, USA) using Odyssey
software. The results were quantified by densitometry (Odyssey
software), normalized for the GAPDH or total AKT levels, and
then the ratio of the experimental values to the control values was
calculated.
Statistical Analysis
Data were expressed as mean 6standard error of the mean
(SEM). Histological data from each animal were obtained by
averaging all values acquired in each tissue section. Statistical
analysis was performed using one-way analysis of variance
(ANOVA) followed by the Bonferroni post hoc test. Confocal
microscopy data were expressed as the percentage of the mean
gray value in relation to the maximum value acquired in the ISO-
treated group of each imaging session and the statistical analysis
was performed using an unpaired Student’s t-test followed by the
Mann-Whitney Utest. One-way ANOVA followed by the
Newman-Keuls post hoc test was used to evaluate the cardiac
function. P,0.05 was considered statistically significant.
Results
The cardiomyocyte cross-sectional area, a measure of cardiac
hypertrophy, was significantly increased in ISO +vehicle-treated
animals compared to oil +vehicle-treated rats (11.1760.09 mm vs.
13.9160.17 mm in ISO +vehicle-treated rats, Figure 1). CGEN-
856S treatment reduced the degree of cardiac hypertrophy, as
evidenced by a significant decrease in the cardiomyocyte diameter
(13.9160.17 mm vs. 12.4160.16 mm in ISO +CGEN-856S-
treated rats, Figure 1). Losartan administration also attenuated the
ISO-induced increase in the cardiomyocyte diameter
(13.9160.17 mm vs. 11.6360.08 mm in ISO +losartan-treated
rats, Figure 1). CGEN-856S or losartan +oil treatment did not
significantly affect the cardiomyocyte diameter.
To investigate the effects of CGEN-856S on cardiac fibrosis, we
evaluated the deposition of cardiac type I and III collagen and
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 8 March 2013 | Volume 8 | Issue 3 | e57757
fibronectin using immunofluorescence labeling and quantitative
confocal microscopy. ISO administration resulted in a significant
increase in the deposition of type I and III collagen and fibronectin
(Figure 2). CGEN-856S treatment significantly reduced the
deposition of type I collagen (60.5765.52% vs. 30.6363.00% in
ISO+CGEN-856S-treated rats, Figures 2A and 2B), type III
collagen (63.5464.84% vs. 45.3562.96% in ISO+CGEN-856S-
treated rats, Figures 2A and 2B), and fibronectin (55.2865.84%
vs. 29.9364.48% in ISO+CGEN-856S-treated rats, Figures 2A
and 2B). Similar effects on cardiac fibrosis were obtained when
ISO-treated rats were administered losartan (Figures 2C and 2D).
The cardioprotective actions of CGEN-856S in the ISO model
prompted us to study its effects on MI. As shown in Figure 3, MI
resulted in reduced systolic tension and decreased velocities of
contraction and relaxation (+dT/dt and -dT/dt, respectively)
when compared to the sham-operated group. CGEN-856S
treatment normalized the systolic tension and attenuated the
decrease in the 6dT/dt induced by MI (Figures 3A, 3C, and 3D).
In addition, CGEN-856S administration produced a slight but
significant increase in the coronary flow when compared with
infarcted rats (Figure 3E). No significant changes were observed in
the diastolic function or heart rate among the groups (Figures 3B
and 3F). The actions of CGEN-856S on the cardiac function of
infarcted rats were similar to those effects observed in infarcted
rats treated with captopril, an ACE inhibitor used as a positive
control (Figure 3). Importantly, CGEN-856S treatment reduced
the infarct area when compared to the vehicle-treated group
(23.6861.94% vs. 15.6863.15% in MI+CGEN-856S-treated
rats). In contrast, captopril did not induce any significant effect
on the size of the infarct area (Figure 4).
In order to ascertain whether CGEN-856S stimulates similar
intracellular pathways as Ang-(1–7), we assessed the effects of
CGEN-856S administration on AKT phosphorylation and the
level of p-AKT and p-eNOS in CHO cells transfected with Mas.
As observed in Figure 5A, transfected CHO cells expressed Mas
while untransfected CHO cells (CHO-K1) did not. After 10 min
of stimulation, Ang-(1–7) and CGEN-856S (10
27
mol/L) signif-
icantly increased the level of p-AKT in CHO-Mas cells (Figure 5B).
On the other hand, administration of 10
27
mol/L of CGEN-856S
or Ang-(1–7) did not affect the level of p-AKT in untransfected
CHO cells (Figure 5C). Of note, 10
29
mol/L of Ang-(1–7) and
CGEN-856S also augmented AKT phosphorylation in CHO-Mas
cells (Figure 5D). Furthermore, we observed that both Ang-(1–7)
and CGEN-856S (10
27
mol/L) significantly increased the level of
p-eNOS Ser1177 (Figure 6A), but not p-eNOS Thr495 (Figure 6B)
in CHO-Mas cells. No significant changes in the level of p-eNOS
Ser1177 were observed in untransfected CHO cells (Figure 6C).
Discussion
The most important findings of the present study were that
CGEN-856S, a novel Mas agonist, attenuated the cardiac damage
induced by ISO treatment and MI. Specifically, CGEN-856S
treatment prevented ISO-induced myocardial hypertrophy and
fibrosis, improved heart function, and reduced myocardial injury
in infarcted rats. CGEN-856S has been described as a specific Mas
agonist with a .1000-fold lower affinity for AT
2
receptors than
Ang II. In addition, no evidence of CGEN-856S binding to AT
1
receptors was observed [21]. Thus, we presented strong evidence
that Mas activation using a specific agonist induces cardioprotec-
tive effects, as demonstrated in 2 distinct models of cardiac failure.
Importantly, we were able to exclude any contribution of AT
2
receptors to these effects, as the affinity of CGEN-856S for AT
2
receptors is quite low.
CGEN-856S was first described as a Mas agonist by Shemesh
et al. in 2008 [20]. The authors showed that this peptide elicits
calcium influx in Mas-transfected CHO cells [20]. Additionally,
we observed that CGEN-856S induces vasorelaxation through an
NO- and Mas-dependent mechanism in rat and mouse aorta rings
[21]. Further evidence of CGEN-856S binding to Mas was
obtained by the observation that the Ang-(1–7) analogue D-Ala
7
-
Ang-(1–7) (A-779) abolishes the vasorelaxant effects of CGEN-
856S and by the effective CGEN-856S-induced displacement of
fluorescent Ang-(1–7) [FAM-Ang-(1–7)] binding in Mas-trans-
fected CHO cells [21]. Additionally, this vasodilative effect of
CGEN-356S was absent in the aortic rings of Mas-knockout mice
[21].
The antihypertrophic effect of Ang-(1–7) has been extensively
reported. Transgenic rats [TGR(A1–7)3292] that possess a 2.5-
fold increase in plasma Ang-(1–7) levels showed attenuated ISO-
induced heart hypertrophy [11]. Tallant et al. [12] observed that
Ang-(1–7) directly inhibits the growth of cultured cardiomyocytes
through Mas activation. The antihypertrophic effects of Ang-(1–7)
on Ang II-treated cardiomyocytes were prevented by N(G)-nitro-l-
arginine methyl ester and 1H-1,2,4oxadiazolo4,2-aquinoxalin-1-
one, suggesting that these effects are mediated by the NO/cyclic
guanosine monophosphate (cGMP) pathway [23]. Furthermore,
the nonpeptide analog of Ang-(1–7), AVE 0991, prevented Ang II-
induced myocardial hypertrophy by putatively inhibiting the
transforming growth factor (TGF)-b1/Smad2 signaling pathway
[18]. In line with these findings, the novel Mas agonist, CGEN-
856S, also reduced cardiomyocyte hypertrophy in rats challenged
with ISO, a b-adrenergic agonist capable of inducing cardiac
remodeling.
Another well-characterized action elicited by Ang-(1–7)/Mas is
its antifibrotic effect. Grobe et al. [16] demonstrated that Ang-(1–
7) selectively prevents collagen deposition in DOCA-salt rats. In
an in vitro study, Ang-(1–7) inhibited collagen formation as
measured by [
3
H]proline incorporation in cardiac fibroblasts of
adult rats and decreased the mRNA expression of various growth
factors, including TGF-1, endothelin-1, and leukemia inhibitory
factor [13]. Nadu et al. [9] reported that the ISO-induced increase
of type I and III collagen and fibronectin deposition observed in
normal rats was attenuated in transgenic rats expressing an Ang-
(1–7)-producing fusion protein [TGR(A1–7)3292]. In addition, it
was reported that these transgenic rats are protected against
cardiac dysfunction and fibrosis and show an attenuated increase
in blood pressure after DOCA-salt treatment [24]. DOCA-salt
[TGR(A1–7)3292] rats showed an important local increase in left
ventricular Ang-(1–7) levels, which might have contributed to the
reduced cardiac dysfunction and fibrotic lesions observed in these
animals [24]. The compound AVE 0991 also decreased ISO-
induced ECM protein deposition [17]. Thus, our current data are
in agreement with previous studies using 2 well-known Mas
agonists, Ang-(1–7) and AVE 0991 [25,26]. These data corrob-
orate the concept that Mas stimulation regulates cardiac
remodeling and indicate that the cardioprotective effects of
CGEN-856S are mediated by Mas activation.
Also, according to previous studies using Ang-(1–7) and the Mas
agonist AVE 0991 [25,26], activation of this receptor by CGEN-
856S elicited significant improvements in cardiac function of
infarcted hearts. CGEN-856S treatment restored the systolic
tension, attenuated the MI-induced decrease in 6dT/dt, and
increased the coronary flow as compared to control infarcted rats.
The beneficial effects of Ang-(1–7)/Mas on cardiac function are
one of the most important actions of ACE2/Ang-(1–7)/Mas axis
activation. This includes both vascular and muscular effects
[7,11,14,15,17]. Thus, the availability of compounds such as
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 9 March 2013 | Volume 8 | Issue 3 | e57757
CGEN-856S that specifically activate Mas represents an important
step toward translating these findings into clinical practice.
It is important to note that we previously demonstrated that
CGEN-856S treatment produced merely a small decrease in the
blood pressure of normotensive Wistar rats. This effect was
observed only after infusion of higher doses of CGEN-856S (30
and 300 ng?kg
21
?min
21
). No significant changes in the heart rate
were noted [21]. Thus, we believe that the functional and
structural cardiac effects induced by CGEN-856S observed in our
current study were not due to decreases in blood pressure.
We found that CGEN-856S stimulates AKT phosphorylation
and increases the level of p-AKT and p-eNOS. These data suggest
that, similarly to Ang-(1–7), CGEN-856S induces its beneficial
effects through Mas activation via an eNOS/AKT-dependent
pathway. Evidence for the involvement of Mas in the actions of
CGEN-856S has been reported previously [21]. This new peptide
induced NO-dependent vasodilation mediated by Mas in rat and
mouse aorta rings [21]. Therefore, we hypothesized that CGEN-
856S could trigger underlying mechanisms related to Mas
stimulation. Consistent with our hypothesis, we observed that
AKT and e-NOS were activated by CGEN-856S. Indeed, it has
been reported that Ang-(1–7) elicits AKT phosphorylation in
endothelial cells and cardiomyocytes [22,27,28]. It is important to
note that high levels of NO/NOS might induce pro-oxidant
effects. When NO reacts with superoxide, it generates the oxidant
anion peroxynitrite (ONOO
2
), which provokes lipid peroxidation,
nitrosation of amino acid residues, and disruption of cell
membranes, cell signaling, and cell survival. Peroxynitrite also
exerts proinflammatory actions [29]. Thus, a complete future
study evaluating the effects of CGEN-856S on the oxidative
balance of cardiomyocytes is warranted.
One may argue that 10
29
mol/L of CGEN-856S significantly
increased AKT phosphorylation (ratio between p-AKT and total
AKT, Figure 5D) but not the level of p-AKT (ratio between p-
AKT and GAPDH, Figure 5B) in Mas-transfected CHO cells.
However, this assertion is not true, as a careful analysis of the p-
AKT level data (Figure 5B) reveals that CGEN-856S treatment
almost doubled the quantity of p-AKT in CHO cells. Thus, we
believe that these data did not achieve statistical significance
merely due to the manner in which the data were organized, i.e.,
both concentrations of CGEN-856S (10
29
mol/L and 10
27
mol/
L) were placed in the same graph (Figure 5B).
Of note, in contrast to Ang-(1–7), CGEN-856S presented no
evidence of ACE inhibitory activity and showed low affinity to
AT
1
and AT
2
receptors [21]. This suggests that the cardioprotec-
tive effects induced by CGEN-856S in these cardiac remodeling
models might be independent of any action on ACE activity or
other angiotensin receptors. Also, it is interesting to note that the
CGEN-856S compound is more stable than Ang-(1–7) [21].
However, all of these possibilities warrant confirmation in cardiac
tissues.
In summary, our current findings demonstrated that CGEN-
856S treatment attenuates ISO and MI-induced heart damage
likely through a mechanism involving the Mas/eNOS/AKT
pathway. These data further support the protective role of Ang-(1–
7) in the cardiovascular system and provide evidence that
stimulation of this GPCR might be a potential therapeutic
approach for cardiovascular diseases.
Author Contributions
Conceived and designed the experiments: SQS MB GC YC APA GR AJF
RASS. Performed the experiments: SQS DI MBLC GABS FDM AABP.
Analyzed the data: SQS DI MBLC GABS FDM AABP MB GC YC APA
GR AJF RASS. Contributed reagents/materials/analysis tools: RASS.
Wrote the paper: SQS AJF RASS.
References
1. Brilla CG, Janicki JS, Weber KT (1991) Impaired diastolic function and
coronary reserve in genetic hypertension. Role of interstitial fibrosis and medial
thickening of intramyocardial coronary arteries. Circ Res 69: 107–115.
2. Deschamps AM, Spinale FG (2005) Matrix modulation and heart failure: new
concepts question old beliefs. Curr Opin Cardiol 20: 211–216.
3. Weber KT (2000) Fibrosis and hypertensive heart disease. Curr Opin Cardiol
15: 264–272.
4. Lijnen PJ, Petrov VV (2003) Role of intracardiac renin-angiotensin-aldosterone
system in extracellular matrix remodeling. Methods Find Exp Clin Pharmacol
25: 541–564.
5. Weber KT, Brilla CG (1991) Pathological hypertrophy and cardiac interstitium.
Fibrosis and renin-angiotensin-aldosterone system. Circulation 83: 1849–1865.
6. Sadoshima J, Izumo S (1993) Molecular characterization of angiotensin II-
induced hypertrophy of cardiac myocytes and hyperplasia of cardiac fibroblasts.
Critical role of the AT1 receptor subtype. Circ Res 73: 413–423.
7. Benter IF, Yousif MH, Anim JT, Cojocel C, Diz DI (2006) Angiotensin-(1–7)
prevents development of severe hypertensi on and end-organ damag e in
spontaneously hypertensive rats treated with L-NAME. Am J Physiol Hear t
Circ Physiol 290: H684–691.
8. Grobe JL, Mecca AP, Lingis M, Shenoy V, Bolton TA, et al. (2007) Prevention
of angiotensin II-induced cardiac remodeling by angiotensin-(1–7). Am J Physiol
Heart Circ Physiol 292: H736–742.
9. Nadu AP, Ferreira AJ, Reudelhuber TL, Bader M, Santos RAS (2008) Reduced
isoproterenol-induced renin-angiotensin changes and extracellular matrix
deposition in hearts of TGR(A1–7)3292 rats. J Am Soc Hypertens 2: 341–348.
10. Santos RAS, Simo˜es e Silva AC, Maric C, Silva DMR, Machado RP, et al.
(2003) Angiotensin-(1–7) is an endogenous ligand for the G protein-coupled
receptor Mas. Proc Natl Acad Sci U S A 100: 8258–8263.
11. Santos RAS, Ferreira AJ, Nadu AP, Braga AN, Almeida AP, et al. (2004)
Expression of an angiotensin-(1–7)-producing fusion protein produces cardio-
protective effects in rats. Physiol Genomics 17: 292–299.
12. Tallant EA, Ferrario CM, Gallagher PE (2005) Angiotensin-(1–7) inhibits
growth of cardiac myocytes through activation of the mas receptor. Am J Physiol
Heart Circ Physiol 289: H1560–1566.
13. Iwata M, Cowling RT, Gurantz D, Moore C, Zhang S, et al. (2005)
Angiotensin-(1–7) binds to specific receptors on cardiac fibroblasts to initiate
antifibrotic and antitrophic effects. Am J Physiol Heart Circ Physiol 289:
H2356–2363.
14. Castro CH, Santos RAS, Ferreira AJ, Bader M, Alenina N, et al. (2006) Effects
of genetic deletion of angiotensin-(1–7) receptor Mas on cardiac function during
ischemia/reperfusion in the isolated perfused mouse heart. Life Sci 80: 264–268.
15. Santos RAS, Castro CH, Gava E, Pinheiro SV, Almeida AP, et al. (2006)
Impairment of in vitro and in vivo heart function in angiotensin-(1–7) receptor
MAS knockout mice. Hypertension 47: 996–1002.
16. Grobe JL, Mecca AP, Mao H, Katovich MJ (2006) Chronic angiotensin-(1–7)
prevents cardiac fibrosis in DOCA-salt model of hypertension. Am J Physiol
Heart Circ Physiol 290: H2417–2423.
17. Ferreira AJ, Oliveira TL, Castro MC, Almeida AP, Castro CH, et al. (2007)
Isoproterenol-i nduced impairment of heart function and remodelin g are
attenuated by the nonpeptide angiotensin-(1–7) analogue AVE 0991. Life Sci
81: 916–923.
18. He JG, Chen SL, Huang YY, Chen YL, Dong YG, et al. (2010) The nonpeptide
AVE0991 attenuates myocardial hypertrophy as induced by angiotensin II
through downregulation of transforming growth factor-beta1/Smad2 expres-
sion. Heart Vessels 25: 438–443.
19. Kliger Y, Gofer E, Wool A, Toporik A, Apatoff A, et al. (2008) Predicting
proteolytic sites in extracellular proteins: only halfway there. Bioinformatics 24:
1049–1055.
20. Shemesh R, Toporik A, Levine Z, Hecht I, Rotman G, et al. (2008) Discovery
and validation of novel peptide agonists for G-protein-coupled receptors. J Biol
Chem 283: 34643–34649.
21. Savergnini SQ, Beiman M, Lautner RQ, de Paula-Carvalho V, Allahdadi K, et
al. (2010) Vascular relaxation, antihypertensive effect, and cardioprotection of
a novel peptide agonist of the Mas Receptor. Hypertension 56: 112–120.
22. Sampaio WO, Santos RAS, Faria-Silva R, Mata Machado LT, Schiffrin EL, et
al. (2007) Angiotensin-(1–7) through receptor Mas mediates endothelial nitric
oxide synthase activation via Akt-dependent pathways. Hypertension 49: 185–
192.
23. Gomes ER, Lara AA, Almeida PW, Guimaraes D, Resende RR, et al. (2010)
Angiotensin-(1–7) prevents cardiomyocyte pathological remodeling through
a nitric oxide/gua nosine 39,59-cyclic mon ophosphate-dependent pathway.
Hypertension 55: 153–160.
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 10 March 2013 | Volume 8 | Issue 3 | e57757
24. Santiago NM, Guimara˜es PS, Sirvente RA, Oliveira LA, Irigoyen MC, et al.
(2010) Lifetime overproduction of circulating Angiotensin-(1–7 ) attenuates
deoxycorticosterone acetate-salt hypertension-induced cardiac dysfunction and
remodeling. Hypertension 55: 889–896.
25. Ferreira AJ, Jacoby BA, Araujo CA, Macedo FA, Silva GAB, et al. (2007) The
nonpeptide angiotensin-(1–7) receptor Mas agonist AVE-0991 attenuates heart
failure induced by myocardial infarction. Am J Physiol Heart Circ Physiol 292:
H1113–1119.
26. Loot AE, Roks AJ, Henning RH, Tio RA, Suurmeijer AJ, et al. (2002)
Angiotensin-(1–7) attenuates the development of heart failure after myocardial
infarction in rats. Circulation 105: 1548–1550.
27. Giani JF, Gironacci MM, Munoz MC, Pena C, Turyn D, et al. (2007)
Angiotensin-(1 7) stimulates the phosphorylation of JAK2, IRS-1 and Akt in rat
heart in vivo: role of the AT1 and Mas receptors. Am J Physiol Heart Circ
Physiol 293: H1154–1163.
28. Dias-Peixoto MF, Santos RA, Gomes ER, Alves MN, Almeida PW, et al. (2008)
Molecular mechanisms involved in the angiotensin-(1–7)/Mas signaling pathway
in cardiomyocytes. Hypertension 52: 542–548.
29. Levonen AL, Patel RP, Brookes P, Go YM, Jo H, et al. (2001) Mechanisms of
cell signaling by nitric oxide and peroxynitrite: from mitochondria to MAP
kinases. Antioxid Redox Signal 3: 215–229.
Cardiac Effects of a Novel Mas Agonist
PLOS ONE | www.plosone.org 11 March 2013 | Volume 8 | Issue 3 | e57757
... These compounds were found to display high specificity for the MasR, without affecting AT 1 R or AT 2 R. Along similar lines, CGEN-856S, another MasR agonist exerted vasorelaxant, anti-arrhythmogenic, antihypertensive, and antifibrotic properties [135]. In addition, it improved cardiac function in a heart failure model [136]. Recently, a steroid hormone, 20-Hydroxyecdysone was found to activate the MasR [137]. ...
Article
Pulmonary fibrosis is a devastating lung disease with multifactorial etiology characterized by alveolar injury, fibroblast proliferation and excessive deposition of extracellular matrix proteins, which progressively results in respiratory failure and death. Accumulating evidence from experimental and clinical studies support a central role of the renin angiotensin aldosterone system (RAAS) in the pathogenesis and progression of idiopathic pulmonary fibrosis. Angiotensin II (Ang II), a key vasoactive peptide of the RAAS mediates pro-inflammatory and pro-fibrotic effects on the lungs, adversely affecting organ function. Recent years have witnessed seminal discoveries in the field of RAAS. Identification of new enzymes, peptides and receptors has led to the development of several novel concepts. Of particular interest is the establishment of a protective axis of the RAAS comprising of Angiotensin converting enzyme 2 (ACE2), Angiotensin-(1-7) [Ang-(1-7)], and the Mas receptor (the ACE2/Ang-(1-7)/Mas axis), and the discovery of a functional role for the Angiotensin type 2 (AT2) receptor. Herein, we shall review our current understanding of the role of RAAS in lung fibrogenesis, provide evidence on the anti-fibrotic actions of the newly recognized RAAS components (the ACE2/Ang-(1-7)/Mas axis and AT2 receptor), discuss potential strategies and translational efforts to convert this new knowledge into effective therapeutics for PF.
... Se han desarrollado dos agonistas de receptores Mas, AVE0991 y CGEN-856S, ambos con efectos reductores de presión arterial en ratas DOCA 14,15 . En otro estudio experimental en ratas, el primer compuesto generó disminución del estrés oxidativo, reducción de hipertrofia ventricular y aumento de la fracción de eyección 16 . ...
Article
Full-text available
Arterial hypertension is one of the biggest public health problems. The research in this area has been relentless and productive, allowing to identify new pathophysiological mechanisms from which new therapeutic options are under development. Despite the recognized efficacy and tolerability of currently available drugs, a high number of patients still do not comply with treatment and maintain inadequate blood pressure levels. This review summarizes the literature about new pharmacological alternatives to treat hypertension. The development state of these new medications ranges from a preclinical state to their clinical use in hypertensive patients. Technological strategies aiming at increasing the compliance with anti-hypertensive medications are also mentioned.
... 31,32 Pretreatment with RSV PLGA NPs and RSV 100 reduced the generation of oxygen radicals and helped in the decrease of these enzymes showing preservation of the structural integrity of myocardium. 33,34 CAT, SOD, GPx, GR, GST, and GSH are free radicalscavenging antioxidant enzymes and are the first-line cellular defense against oxidative stress. These enzymes eliminate oxygen radicals such as superoxide (O 2-) and hydrogen peroxide (H 2 O 2 ) and prevent the formation of a more reactive hydroxyl radical (%OH). ...
Article
Full-text available
Our study is aimed at evaluating the effects of pretreatment with Poly(lactic‐co‐glycolic) acid nanoparticle loaded with resveratrol (RSV PLGA NPs) compared to conventional resveratrol (RSV) on isoproterenol (ISO) induced myocardial infarction (MI) in rats. Sixty rats were randomly divided into six groups of 10 rats each. RSV and RSV PLGA NPs were given by gavage in two different doses (50 mg/kg body weight [BW] and 100 mg/kg BW) for 3 weeks. RSV and RSV PLGA NPs were given for 2 weeks starting 1 week before ISO administration. The blood samples were taken 24 hr after the last dose of ISO. The antioxidant, anti‐inflammatory, and cardioprotective effects were evaluated in all groups. Only 100 mg/kg dose of RSV and both doses of RSV PLGA NPs offered a cardioprotective effect by preventing cardiac troponin T (cTnT) levels, lactate dehydrogenase (LDH), and aspartate aminotransferase (AST) activities leakage from cardiomyocytes, with the best result for RSV PLGA NPs. All the oxidative stress parameters were significantly improved after RSV PLGA NPs compared to RSV pretreatment. RSV PLGA NPs were more efficient than RSV in limiting the increase in inflammatory cytokine expressions such as tumor necrosis factor‐alpha (TNF‐α), interleukin 1 beta (IL‐1β), and NF‐kappaB (NF‐kB) expression. In addition, RSV PLGA NPs significantly upregulated eNOS expression and downregulated iNOS expression. RSV PLGA NPs better prevented myocardial necrosis and reduced interstitial edema and neutrophil infiltration than RSV, on histopathological examination. Therefore, improving the bioactivity of RSV by nanotechnology may help limit cardiac injury after myocardial infarction.
... 73 MasR agonism, with either AVE0991 or CGEN-856, also prevents myocardial infarction-related injury remodeling, and attenuates myocardial hypertrophy in rats. 25,74 Recent efforts to improve the pharmacokinetic profile and therapeutic potential have led to testing of Ang-(1-7) encapsulation by hydroxyl-propyl-b-cyclodextrin (HPβCD) to prevent rapid degradation and improve oral bioavailability. HPβCD-Ang-(1-7) reduces blood pressure and heart rate, improves cardiac function and fibrosis, and promotes antithrombotic effects in rat models of hypertension and myocardial infarction. ...
Article
Despite decades of research and numerous treatment approaches, hypertension and cardiovascular disease remain leading global public health problems. A major contributor to regulation of blood pressure, and the development of hypertension, is the renin-angiotensin system. Of particular concern, uncontrolled activation of angiotensin II contributes to hypertension and associated cardiovascular risk, with antihypertensive therapies currently available to block the formation and deleterious actions of this hormone. More recently, angiotensin-(1-7) has emerged as a biologically active intermediate of the vasodilatory arm of the renin-angiotensin system. This hormone antagonizes angiotensin II actions as well as offers antihypertensive, antihypertrophic, antiatherogenic, antiarrhythmogenic, antifibrotic and antithrombotic properties. Angiotensin-(1-7) elicits beneficial cardiovascular actions through mas G protein-coupled receptors, which are found in numerous tissues pivotal to control of blood pressure including the brain, heart, kidneys, and vasculature. Despite accumulating evidence for favorable effects of angiotensin-(1-7) in animal models, there is a paucity of clinical studies and pharmacokinetic limitations, thus limiting the development of therapeutic agents to better understand cardiovascular actions of this vasodilatory peptide hormone in humans. This review highlights current knowledge on the role of angiotensin-(1-7) in cardiovascular control, with an emphasis on significant animal, human, and therapeutic research efforts.
Article
MasR is a critical element in the RAS accessory pathway that protects the heart against myocardial infarction, ischemia-reperfusion injury, and pathological remodeling by counteracting the effects of AT1R. This receptor is mainly stimulated by Ang 1-7, which is a bioactive metabolite of the angiotensin produced by ACE2. MasR activation attenuates ischemia-related myocardial damage by facilitating vasorelaxation, improving cell metabolism, reducing inflammation and oxidative stress, inhibiting thrombosis, and stabilizing atherosclerotic plaque. It also prevents pathological cardiac remodeling by suppressing hypertrophy- and fibrosis-inducing signals. In addition, the potential of MasR in lowering blood pressure, improving blood glucose and lipid profiles, and weight loss has made it effective in modulating risk factors for coronary artery disease including hypertension, diabetes, dyslipidemia, and obesity. Considering these properties, the administration of MasR agonists offers a promising approach to the prevention and treatment of ischemic heart disease.Abbreviations: Acetylcholine (Ach); AMP-activated protein kinase (AMPK); Angiotensin (Ang); Angiotensin receptor (ATR); Angiotensin receptor blocker (ARB); Angiotensin-converting enzyme (ACE); Angiotensin-converting enzyme inhibitor (ACEI); Anti-PRD1-BF1-RIZ1 homologous domain containing 16 (PRDM16); bradykinin (BK); Calcineurin (CaN); cAMP-response element binding protein (CREB); Catalase (CAT); C-C Motif Chemokine Ligand 2 (CCL2); Chloride channel 3 (CIC3); c-Jun N-terminal kinases (JNK); Cluster of differentiation 36 (CD36); Cocaine- and amphetamine-regulated transcript (CART); Connective tissue growth factor (CTGF); Coronary artery disease (CAD); Creatine phosphokinase (CPK); C-X-C motif chemokine ligand 10 (CXCL10); Cystic fibrosis transmembrane conductance regulator (CFTR); Endothelial nitric oxide synthase (eNOS); Extracellular signal-regulated kinase 1/2 (ERK 1/2); Fatty acid transport protein (FATP); Fibroblast growth factor 21 (FGF21); Forkhead box protein O1 (FoxO1); Glucokinase (Gk); Glucose transporter (GLUT); Glycogen synthase kinase 3β (GSK3β); High density lipoprotein (HDL); High sensitive C-reactive protein (hs-CRP); Inositol trisphosphate (IP3); Interleukin (IL); Ischemic heart disease (IHD); Janus kinase (JAK); Kruppel-like factor 4 (KLF4); Lactate dehydrogenase (LDH); Left ventricular end-diastolic pressure (LVEDP); Left ventricular end-systolic pressure (LVESP); Lipoprotein lipase (LPL); L-NG-Nitro arginine methyl ester (L-NAME); Low density lipoprotein (LDL); Mammalian target of rapamycin (mTOR); Mas-related G protein-coupled receptors (Mrgpr); Matrix metalloproteinase (MMP); MAPK phosphatase-1 (MKP-1); Mitogen-activated protein kinase (MAPK); Monocyte chemoattractant protein-1 (MCP-1); NADPH oxidase (NOX); Neuropeptide FF (NPFF); Neutral endopeptidase (NEP); Nitric oxide (NO); Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB); Nuclear-factor of activated T-cells (NFAT); Pancreatic and duodenal homeobox 1 (Pdx1); Peroxisome proliferator- activated receptor γ (PPARγ); Phosphoinositide 3-kinases (PI3k); Phospholipase C (PLC); Prepro-orexin (PPO); Prolyl-endopeptidase (PEP); Prostacyclin (PGI2); Protein kinase B (Akt); Reactive oxygen species (ROS); Renin-angiotensin system (RAS); Rho-associated protein kinase (ROCK); Serum amyloid A (SAA); Signal transducer and activator of transcription (STAT); Sirtuin 1 (Sirt1); Slit guidance ligand 3 (Slit3); Smooth muscle 22α (SM22α); Sterol regulatory element-binding protein 1 (SREBP-1c); Stromal-derived factor-1a (SDF); Superoxide dismutase (SOD); Thiobarbituric acid reactive substances (TBARS); Tissue factor (TF); Toll-like receptor 4 (TLR4); Transforming growth factor β1 (TGF-β1); Tumor necrosis factor α (TNF-α); Uncoupling protein 1 (UCP1); Ventrolateral medulla (VLM).
Article
The renin–angiotensin system (RAS) has currently attracted increasing attention due to its potential function in regulating energy homeostasis, other than the actions on cellular growth, blood pressure, fluid, and electrolyte balance. The existence of RAS is well established in metabolic organs, including pancreas, liver, skeletal muscle, and adipose tissue, where activation of angiotensin-converting enzyme (ACE) – angiotensin II pathway contributes to the impairment of insulin secretion, glucose transport, fat distribution, and adipokines production. However, the activation of angiotensin-converting enzyme 2 (ACE2) – angiotensin (1–7) pathway, a novel branch of the RAS, plays an opposite role in the ACE pathway, which could reverse these consequences by improving local microcirculation, inflammation, stress state, structure remolding, and insulin signaling pathway. In addition, new studies indicate the protective RAS arm possesses extraordinary ability to enhance brown adipose tissue (BAT) activity and induces browning of white adipose tissue, and consequently, it leads to increased energy expenditure in the form of heat instead of ATP synthesis. Interestingly, ACE2 is the receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is threating public health worldwide. The main complications of SARS-CoV-2 infected death patients include many energy metabolism-related chronic diseases, such as diabetes. The specific mechanism leading to this phenomenon is largely unknown. Here, we summarize the latest pharmacological and genetic tools on regulating ACE/ACE2 balance and highlight the beneficial effects of the ACE2 pathway axis hyperactivity on glycolipid metabolism, as well as the thermogenic modulation.
Article
Full-text available
We evaluated the development of arterial hypertension, cardiac function, and collagen deposition, as well as the level of components of the renin-angiotensin system in the heart of transgenic rats that overexpress an angiotensin (Ang)-(1-7)-producing fusion protein, TGR(A1-7)3292 (TG), which induces a lifetime increase in circulating levels of this peptide. After 30 days of the induction of the deoxycorticosterone acetate (DOCA)-salt hypertension model, DOCA-TG rats were hypertensive but presented a lower systolic arterial pressure in comparison with DOCA-Sprague-Dawley (SD) rats. In contrast to DOCA-SD rats that presented left ventricle (LV) hypertrophy and diastolic dysfunction, DOCA-TG rats did not develop cardiac hypertrophy or changes in ventricular function. In addition, DOCA-TG rats showed attenuation in mRNA expression for collagen type I and III compared with the increased levels of DOCA-SD rats. Ang II plasma and LV levels were reduced in SD and TG hypertensive rats in comparison with normotensive animals. DOCA-TG rats presented a reduction in plasma Ang-(1-7) levels; however, there was a great increase in Ang-(1-7) ( approximately 3-fold) accompanied by a decrease in mRNA expression of both angiotensin-converting enzyme and angiotensin-converting enzyme 2 in the LV. The mRNA expression of Mas and Ang II type 1 receptors in the LV was not significantly changed in DOCA-SD or DOCA-TG rats. This study showed that TG rats with increased circulating levels of Ang-(1-7) are protected against cardiac dysfunction and fibrosis and also present an attenuated increase in blood pressure after DOCA-salt hypertension. In addition, DOCA-TG rats showed an important local increase in Ang-(1-7) levels in the LV, which might have contributed to the attenuation of cardiac dysfunction and prefibrotic lesions.
Article
Full-text available
G-protein-coupled receptors (GPCRs) represent an important group of targets for pharmaceutical therapeutics. The completion of the human genome revealed a large number of putative GPCRs. However, the identification of their natural ligands, and especially peptides, suffers from low discovery rates, thus impeding development of therapeutics based on these potential drug targets. We describe the discovery of novel GPCR ligands encrypted in the human proteome. Hundreds of potential peptide ligands were predicted by machine learning algorithms. In vitro screening of selected 33 peptides on a set of 152 GPCRs, including a group of designated orphan receptors, was conducted by intracellular calcium measurements and cAMP assays. The screening revealed eight novel peptides as potential agonists that specifically activated six different receptors in a dose-dependent manner. Most of the peptides showed distinct stimulatory patterns targeted at designated and orphan GPCRs. Further analysis demonstrated a significant in vivo effect for one of the peptides in a mouse inflammation model.
Article
Background— The renin-angiotensin system (RAS) is a key player in the progression of heart failure. Angiotensin-(1–7) is thought to modulate the activity of the RAS. Furthermore, this peptide may play a part in the beneficial effects of angiotensin-converting enzyme inhibitors in cardiovascular disease. We assessed the effects of angiotensin-(1–7) on the progression of heart failure. Methods and Results— Male Sprague-Dawley rats underwent either coronary ligation or sham surgery. Two weeks after induction of myocardial infarction, intravenous infusion of angiotensin-(1–7) (24 μg/kg per hour) or saline was started by minipump. After 8 weeks of treatment, hemodynamic parameters were measured, endothelial function was assessed in isolated aortic rings, and plasma angiotensin-(1–7) levels were determined. Myocardial infarction resulted in a significant deterioration of left ventricular systolic and diastolic pressure, dP/dt, and coronary flow. Raising plasma levels 40-fold, angiotensin-(1–7) infusion attenuated this impairment to a nonsignificant level, markedly illustrated by a 40% reduction in left ventricular end-diastolic pressure. Furthermore, angiotensin-(1–7) completely preserved aortic endothelial function, whereas endothelium-dependent relaxation in aortas of saline-treated infarcted rats was significantly decreased. Conclusions— Angiotensin-(1–7) preserved cardiac function, coronary perfusion, and aortic endothelial function in a rat model for heart failure.
Article
Functional angiotensin II receptors have been documented in cardiac fibroblasts as well as an intracardiac aldosterone system that responds to short- and long-term physiological stimuli. In vitro, angiotensin II increased cardiac fibroblast-mediated collagen synthesis and mRNA levels of collagen type I, type III, pro-alpha1 (I) collagen, pro-alpha1 (III) collagen and fibronectin, and inhibited matrix metalloproteinase I activity. The angiotensin II-stimulated secretion and expression of collagen was completely abolished by AT1 receptor antagonism, but not affected by AT2 receptor antagonism. In vivo, chronic infusion of angiotensin II increased the collagen volume fraction in the ventricles. Angiotensin-converting enzyme (ACE) inhibition and AT1 receptor antagonism, but not AT2 receptor antagonism, reduced collagen deposition in the myocardium in spontaneously hypertensive rats and in rat myocardium following myocardial infarction. During chronic aldosterone infusion in uninephrectomized rats on a high-salt diet, a marked accumulation of interstitial and to a lesser extent perivascular collagen occurs in the heart in both ventricles. The cardiac fibrosis in this aldosterone model is prevented by spironolactone. During the continuous infusion of aldosterone in the rat, the appearance of fibrosis was delayed and started 4 weeks after the beginning of the infusion, which argues against a direct effect of aldosterone. The mechanism of aldosterone-salt-induced cardiac fibrosis possibly involves angiotensin II acting through upregulated AT1 receptors and the cardiac AT1 receptor is the target for aldosterone. An accumulation of collagen in the heart has also been found in patients with adrenal adenomas and during chronic activation of the renin-angiotensin-aldosterone system such as in surgically-induced unilateral renal ischemia, unilateral renal artery banding or renovascular hypertension. Spironolactone prevents aortic collagen accumulation in spontaneously hypertensive rats. In patients with stable chronic heart failure, spironolactone treatment in addition to diuretics and ACE inhibition reduced circulating levels of procollagen type III N-terminal aminopeptide. Also, in the Randomized Aldactone Evaluation Study, spironolactone coadministered with conventional therapy of ACE inhibitors, loop diuretics and digitalis in patients with symptomatic heart failure defined as NYHA classes III-IV, reduced total mortality by 30%.
Article
The nonpeptide AVE0991 is expected to be a putative new drug for cardiovascular diseases. However, the mechanisms for the cardioprotective actions of AVE0991 are still not fully understood. We planned to determine whether AVE0991 attenuates the angiotensin II (AngII)-induced myocardial hypertrophy and whether these AVE0991 effects involved transforming growth factor beta1 (TGF-beta1) and Smad2. A rat model of neonatal myocardial hypertrophy was induced by AngII. The AngII group significantly increased in protein content, surface area, and [(3)H]leucine incorporation efficiency by cardiomyocytes, compared to those of the control group (P < 0.01). The AngII group also had elevated TGF-beta1 and Smad2 expression (P < 0.01). These AngII-induced changes were significantly attenuated by AVE0991 in a dose-dependent manner. In our study, these actions of AngII (10(-6) mol/l) were significantly inhibited by both concentrations of AVE0991 (10(-5) mol/l and 10(-7) mol/l). Moreover, the high AVE0991 group had significantly better inhibition of myocardial hypertrophy than the low AVE0991 group. Meanwhile, the beneficial effects of AVE0991 were completely abolished when the cardiomyocytes were pretreated with Ang-(1-7) receptor antagonist A-779 (10(-6) mol/l). These results suggested that AVE0991 prevented AngII-inducing myocardial hypertrophy in a dose-dependent fashion, a process that may be associated with the inhibition of TGF-beta1/Smad2 signaling.
Article
Mas stimulation with angiotensin (Ang)-(1-7) produces cardioprotective effects and vasorelaxation. Using a computational discovery platform for predicting novel naturally occurring peptides that may activate G protein-coupled receptors, we discovered a novel Mas agonist peptide, CGEN-856S. An endothelium- and NO-dependent vasodilating effect was observed for CGEN-856S in thoracic aorta rings of rats (maximal value for the relaxant effect: 39.99+/-5.034%), which was similar to that produced by Ang-(1-7) (10(-10) to 10(-6) mol/L). In addition, the vasodilator activity of this peptide depended on a functional Mas receptor, because it was abolished in aorta rings of Mas-knockout mice. CGEN-856S appears to bind the Mas receptor at the same binding domain as Ang-(1-7), as suggested by the blocking of its vasorelaxant effect with the Ang-(1-7) analogue d-Ala(7)-Ang-(1-7), and by its competitive inhibition of Ang-(1-7) binding to Mas-transfected cells. The effect of CGEN-856S on reperfusion arrhythmias and cardiac function was studied on ischemia reperfusion of isolated rat hearts. We found that picomolar concentration of CGEN-856S (0.04 nmol/L) had an antiarrhythmogenic effect, as demonstrated by a reduction in the incidence and duration of reperfusion arrhythmias. Furthermore, acute infusion of CGEN-856S produced a shallow dose-dependent decrease in mean arterial pressure of conscious spontaneously hypertensive rats. The maximum change during infusion was observed at the highest dose. Strikingly, blood pressure continued to drop in the postinfusion period. The results presented here indicate that the novel Mas agonist, CGEN-856S, might have a therapeutic value, because it induces vasorelaxing, antihypertensive, and cardioprotective effects.
Article
We investigated the expression of specific extracellular matrix (ECM) proteins in cardiac hypertrophy induced by isoproterenol in TGR(A1-7)3292 rats. Additionally, changes in circulating and tissue renin-angiotensin system (RAS) were analyzed. Left ventricles (LV) were used for quantification of collagen type I, III, and fibronectin using immunofluorescence-labeling techniques. Angiotensin (Ang) II levels were measured by radioimmunoassay. Expression of RAS components was assessed by semi-quantitative polymerase chain reaction (PCR) or real-time PCR. Isoproterenol treatment induced an increase in the expression of collagen I, III, and fibronectin in normal rats. Collagen I and fibronectin expression were decreased in TGR(A1-7)3292 at basal conditions and both proteins increased by isoproterenol treatment; however, the levels achieved were still significantly lower than those observed in treated normal rats. The increase in collagen III observed in normal rats was completely blunted in TGR(A1-7)3292. Moreover, TGR(A1-7)3292 presented lower Ang II levels and angiotensinogen expression and a higher angiotensin-converting enzyme 2 (ACE2) expression in LV. Isoproterenol treatment increased cardiac Ang II concentration only in normal rats, which was associated with an increase in ACE2 and a decrease in Mas expression. These observations suggest that Ang-(1-7) specifically modulates the expression of RAS components and ECM proteins in LV.
Article
The renin-angiotensin (Ang) system plays a pivotal role in the pathogenesis of cardiovascular disease, with Ang II being the major effector of this system. Multiple lines of evidence have shown that Ang-(1-7) exerts cardioprotective effects in the heart by counterregulating Ang II actions. The questions that remain are how and where Ang-(1-7) exerts its effects. By using a combination of molecular biology, confocal microscopy, and a transgenic rat model with increased levels of circulating Ang-(1-7) (TGR[A1-7]3292), we evaluated the signaling pathways involved in Ang-(1-7) cardioprotection against Ang II-induced pathological remodeling in ventricular cardiomyocytes. Rats were infused with Ang II for 2 weeks. We found that ventricular myocytes from TGR(A1-7)3292 rats are protected from Ang II pathological remodeling characterized by Ca(2+) signaling dysfunction, hypertrophic fetal gene expression, glycogen synthase kinase 3beta inactivation, and nuclear factor of activated T-cells nuclear accumulation. Moreover, cardiomyocytes from TGR(A1-7)3292 rats infused with Ang II presented increased expression levels of neuronal NO synthase. To provide a signaling pathway involved in the beneficial effects of Ang-(1-7), we treated neonatal cardiomyocytes with Ang-(1-7) and Ang II for 36 hours. Treatment of cardiomyocytes with Ang-(1-7) prevented Ang II-induced hypertrophy by modulating calcineurin/nuclear factor of activated T-cell signaling cascade. Importantly, antihypertrophic effects of Ang-(1-7) on Ang II-treated cardiomyocytes were prevented by N(G)-nitro-l-arginine methyl ester and 1H-1,2,4oxadiazolo4,2-aquinoxalin-1-one, suggesting that these effects are mediated by NO/cGMP. Taken together, these data reveal a key role for NO/cGMP as a mediator of Ang-(1-7) beneficial effects in cardiac cells.
Article
Recently there has been growing evidence suggesting that beneficial effects of angiotensin-(1-7) [Ang-(1-7)] in the heart are mediated by its receptor Mas. However, the signaling pathways involved in these effects in cardiomyocytes are unknown. Here, we investigated the involvement of the Ang-(1-7)/Mas axis in NO generation and Ca(2+) handling in adult ventricular myocytes using a combination of molecular biology, intracellular Ca(2+) imaging, and confocal microscopy. Acute Ang-(1-7) treatment (10 nmol/L) leads to NO production and activates endothelial NO synthase and Akt in cardiomyocytes. Ang-(1-7)-dependent NO raise was abolished by pretreatment with A-779 (1 micromol/L). To confirm that Ang-(1-7) action is mediated by Mas, we used cardiomyocytes isolated from Mas-deficient mice. In Mas-deficient cardiomyocytes, Ang-(1-7) failed to increase NO levels. Moreover, Mas-ablation was accompanied by significant alterations in the proteins involved in the regulation of endothelial NO synthase activity, indicating that endothelial NO synthase and its binding partners are important effectors of the Mas-mediated pathway in cardiomyocytes. We then investigated the role of the Ang-(1-7)/Mas axis on Ca(2+) signaling. Cardiomyocytes treated with 10 nmol/L of Ang-(1-7) did not show changes in Ca(2+)-transient parameters such as peak Ca(2+) transients and kinetics of decay. Nevertheless, cardiomyocytes from Mas-deficient mice presented reduced peak and slower [Ca(2+)](i) transients when compared with wild-type cardiomyocytes. Lower Ca(2+) ATPase of the sarcoplasmic reticulum expression levels accompanied the reduced Ca(2+) transient in Mas-deficient cardiomyocytes. Therefore, chronic Mas-deficiency leads to impaired Ca(2+) handling in cardiomyocytes. Collectively, these observations reveal a key role for the Ang-(1-7)/Mas axis as a modulator of cardiomyocyte function.