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

Current Hypertension Reports (2018) 20: 17

https://doi.org/10.1007/s11906-018-0798-6

PATHOGENESIS OF HYPERTENSION (W ELLIOTT AND R SANTOS, SECTION EDITORS)

Angiotensin-(1–7) and Alamandine on Experimental Models


of Hypertension and Atherosclerosis
Fernando Pedro de Souza-Neto 1 & Melissa Carvalho Santuchi 1 & Mario de Morais e Silva 1 &
Maria José Campagnole-Santos 1 & Rafaela Fernandes da Silva 1

Published online: 14 March 2018


# Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Purpose of Review The purpose of this review was to summarize the current knowledge on the role of angiotensin-(1–7)
[Ang-(1–7)] and alamandine in experimental hypertension and atherosclerosis.
Recent Findings The renin-angiotensin system (RAS) is a very complex system, composed of a cascade of enzymes, peptides,
and receptors, known to be involved in the pathogenesis of hypertension and atherosclerosis. Ang-(1–7), identified and charac-
terized in 1987, and alamandine, discovered 16 years after, are the newest two main effector molecules from the RAS, protecting
the vascular system against hypertension and atherosclerosis.
Summary While the beneficial effects of Ang-(1–7) have been widely studied in several experimental models of hypertension,
much less studies were performed in experimental models of atherosclerosis. Alamandine has shown similar vascular effects to
Ang-(1–7), namely, endothelial-dependent vasorelaxation mediated by nitric oxide and hypotensive effects in experimental
hypertension. There are few studies on the effects of alamandine on atherosclerosis.

Keywords Renin-angiotensin system . Angiotensin-(1–7) . Mas receptor . Alamandine . MrgD receptor; hypertension .
Atherosclerosis

Introduction muscle cell (VSMC) phenotype, presence of vascular inflam-


mation and oxidative stress, and activation of the renin-
There is a global epidemia of cardiovascular disease (CVD). angiotensin system (RAS) [2, 3].
The number of cases of CVD increased to 17.3 million in RAS is a very complex and dynamic system, composed of
2013, according to the Global Burden of Disease Study a cascade of enzymes and peptides, important for many phys-
2013 [1]. Hypertension and atherosclerosis are major risk fac- iological processes and involved in the pathogenesis of hyper-
tors for CVD morbidity and mortality. Although considered as tension and atherosclerosis [4–6]. Initially described as a hor-
independent risk factors, they are closely related and very monal system, it is now well known that many of the enzymes
often occur simultaneously, affecting the cardiovascular sys- and peptides can be produced locally in a variety of organs,
tem and resulting in damage to various organs. Hypertension including blood vessels. Angiotensin II (Ang II) is the best-
and atherosclerosis share some common vascular events such studied peptide, considered as the major end effector molecule
as endothelium dysfunction, alterations on vascular smooth of the classical RAS axis, composed of angiotensin-
converting enzyme (ACE), Ang II, and angiotensin receptor
This article is part of the Topical Collection on Pathogenesis of type I (AT1R). The activation of ACE/Ang II/AT1R axis
Hypertension participates on blood pressure increase, as many studies
have demonstrated that chronic pharmacological administra-
* Rafaela Fernandes da Silva tion of ACE inhibitors and AT1R blockers lowers blood
rfdasilva.ufmg@gmail.com
pressure in several animal models of hypertension and in
1 humans [7]. Ang II generated in the brain, kidney, and blood
Department of Physiology and Biophysics, Institute of Biological
Sciences, Federal University of Minas Gerais, Av. Antônio Carlos. vessels promotes hypertension involving a complex mecha-
6627, Pampulha, Belo Horizonte, MG 31270-901, Brazil nism, including transcriptional activation, reactive oxygen
17 Page 2 of 15 Curr Hypertens Rep (2018) 20: 17

species (ROS) production, inflammation, and many other Renin (30–40 kDa), a monospecific aspartyl protease, is
cellular events. Ang II promotes atherosclerosis and partici- synthesized, stored, and released in its active form or as
pates in all phases of the disease, from the early lesion prorenin from juxtaglomerular cells in the kidneys [14].
formation, growth, progression, until plaque rupture [8, 9]. Many studies have shown that renin can also be synthesized
Most of the mechanisms described about the participation of in the collecting tubules and released into tubular fluid [15,
Ang II on atherosclerosis involve oxidative stress and in- 16]. In addition, there is evidence for intracellular production
flammation [10]. of renin in the brain, heart, and adrenals [15, 17]. Interestingly,
In the last decades, new RAS components have been dis- prorenin has been identified as a hormonal component of
covered and characterized, changing the classical view of RAS, acting on receptors for prorenin in various tissues [18].
RAS as a linear cascade. The existence of an alternative axis, Ang I is a decapeptide with very weak biological activity
composed by angiotensin-converting enzyme 2 (ACE2), and serves as a precursor for the formation of Ang II, an
Ang-(1–7), and Mas receptor (MasR), that counteract many octapeptide hormone considered the main effector of RAS
of the effects of ACE/Ang II/AT1R is now well accepted [4]. [19]. ACE (150–180 kDa) cleaves the dipeptide histidyl-
Indeed, the activation of ACE2/Ang-(1–7)/MasR by pharma- leucine in the C-terminal portion of Ang II [20–22].
cological treatments or by genetic modification has shown Chymase (35 kDa) is a peptidase that can also form Ang II
antihypertensive and antiatherosclerotic effects in different by hydrolyzing the Phe8-His9 bond of Ang I, in addition to
experimental disease models. being capable of metabolizing Ang II itself and forming
Finally, in 2013 the research group of Santos identified and Ang-(1–4) and Ang-(1–5) [23]. Ang II is the main active mol-
characterized the newest peptide of the RAS. The ecule from the classical RAS, interacting with AT1R and an-
heptapeptide Ala1-Ang-(1–7) was detected in plasma of rats giotensin receptor type II (AT2R) in blood vessels, heart, kid-
and humans, and in the aorta of mice and rats. Named as neys, and brain. The inappropriate overactivity of the Ang II/
alamandine, the chemical structure of this peptide is very sim- AT1R interaction is involved in the genesis and progression of
ilar to Ang-(1–7), with only one amino acid residue differ- hypertension and atherosclerosis [24–26].
ence, Ala1, in the N-terminal residue instead of Asp1 [11••]. The understanding of RAS has been expanded, as new
In this pioneering discovery article, Lautner, Vilella, Fraga- components have been discovered and characterized, such as
Silva et al. [11••] showed that alamandine, differently from ACE2, Ang-(1–7) and MasR. In 1988, Santos and colleagues
Ang-(1–7), acts preferentially by its binding to the MrgD re- showed for the first time the formation of Ang-(1–7) after the
ceptor [11••]. In spite of that, alamandine presented similar incubation of dog brainstem homogenates with [125I]-Ang I,
hypotensive effects to Ang-(1–7) in conscious spontaneously in the absence or presence of ACE inhibitors [27••]. Thus,
hypertensive rats (SHR) and when microinjected into the cau- these authors demonstrated that Ang-(1–7) was not only
dal ventrolateral medulla of anesthetized Fisher rats. formed by the metabolism of Ang II. In this same year,
Thereafter, two additional studies confirmed the hypotensive Schiavone et al. [28] showed that Ang-(1–7) was able to in-
effects of alamandine. In regard to atherosclerosis, up to the duce neurosecretory activity in the rat hypothalamo-
present, only two studies have been published, with distinct neurohypophysial system (HNS), promoting release of arginin
outcomes that it will be further discussed in this review. vasopressin (AVP) in vitro. Campagnole-Santos et al. [29]
Thus, the aim of this review article is to summarize the also demonstrated that Ang-(1–7) promotes hypotensive re-
current knowledge about the role of the two major peptides sponses when microinjected into the dorsal medulla of rats,
effectors from the novel protective RAS axes, Ang-(1–7) and more specifically in the nucleus of the solitary tract. In addi-
alamandine, in experimental hypertension and atherosclerosis. tion to these evidences of the biological activity of Ang-(1–7)
in the nervous system, Chappel et al. identified the presence of
Ang-(1–7) in rat blood circulation and adrenals [30••].
Novel RAS Axes Endogenous Ang-(1–7) can be formed directly from Ang I
by the hydrolysis of the Pro7-Phe8 peptide bond, in the C-
RAS is an important endocrine, paracrine, and autocrine sys- terminal portion by the enzymes prolyl oligopeptidase and
tem responsible for the control of blood pressure, thimet oligopeptidase, besides neprilysin. In addition, the ac-
hydroelectrolyte balance, and maintenance of cardiovascular tion of ACE on Ang I produces Ang II, which subsequently
and renal structures and functions [9]. The classical axis of this undergoes enzymatic action of ACE2 on the residue of Phe8 in
system is initiated with angiotensinogen, a 411-amino acid its C-terminal moiety to give rise to Ang-(1–7) [31]. ACE2
(50–60 kDa) 2-globulin, synthesized in the liver from a single can also act on Ang I to form Ang-(1–9), which in turn is
gene and released into the bloodstream or produced at tissue converted to Ang-(1–7) [32].
level [12]. This macromolecule is the only precursor to all In the year of 2003, Santos et al. identified Mas as the
components of RAS and is cleaved by renin-forming angio- functional receptor for Ang-(1–7). In this study, they demon-
tensin I (Ang I) from its N-terminal portion [13]. strated that the antidiuretic effect elicited by Ang-(1–7) was
Curr Hypertens Rep (2018) 20: 17 Page 3 of 15 17

abolished in MasR-knockout mice. It was also observed that and how they play a role in the pathophysiology of cardiovas-
Ang-(1–7) promotes the release of arachidonic acid in cells cular diseases. Here, we will highlight the main results obtained
transfected with this receptor, but the use of D-Ala7-Ang-(1–7) with the use of alamandine in experimental hypertension and
(A-779), a potent and selective MasR antagonist, blocks this atherosclerosis.
effect. Furthermore, dose-dependent relaxation of Ang-(1–7)
was abolished in MasR-deficient aortic rings, without interfering
with endothelium-dependent relaxation by acetylcholine [33••]. Angiotensin-(1–7) and Hypertension
Different studies have shown that activation of the
ACE2/Ang-(1–7)/MasR axis counteracts with many of the car- The axis formed by ACE2/Ang-(1–7)/MasR is an important
diovascular effects of ACE/Ang II/AT1R, including anti-inflam- regulator of cardiovascular functions. Its effects on blood pres-
matory, antiproliferative, antifibrotic, antihypertrophic, and sure occur mainly through its direct actions in blood vessels,
vasodilatory mechanisms of action [34–36]. kidneys, and brain (Fig. 1) [39]. Administration of Ang-(1–7)
Other peptides are also formed within the RAS and produce in adult Akita mice (type I spontaneously diabetic animals)
biological effects, such as angiotensin-(2–8) (Ang III) and an- over a period of 6 weeks was able to protect against elevated
giotensin-(3–8) (Ang IV) [34]. In addition to these, other pep- systolic blood pressure (SBP), which was abolished by the
tides have been identified more recently, such as angiotensin A simultaneous administration of A-779 [40]. Kangussu et al.
(Ang A), a vasoconstrictor octapeptide similar to Ang II, that demonstrated that intracerebroventricular administration of
exerts its effects through AT1R [37] and alamandine that can be Ang-(1–7) reduced systolic and diastolic blood pressure in
formed by the action of ACE2 on the octapeptide angiotensin A transgenic hypertensive rats (mRen2)27 [41]. In the same
or by thedecarboxylation of the N-terminal amino acid of study, it was demonstrated that Ang-(1–7) also improves car-
Ang-(1–7), Asp1, into Ala1 [11]. Alamandine can be formed diac parameters related to the hypertension, such as reduction
by the action of ACE2 on the octapeptide Ang A or by the of cardiomyocyte hypertrophy, total collagen deposition, and
decarboxylation of the N-terminal amino acid of Ang-(1–7), type I collagen and fibronectin gene expressions, as well as
Asp1, into Ala1. Although the molecular difference between reduction of the ratio between cardiac sympathetic and para-
Ang-(1–7) and alamandine is only one amino acid residue, it sympathetic tonus. Fall in blood pressure was also observed in
was shown that the vasorelaxation and hypotensive effects of transgenic mice overexpressing human ACE2 selectively in the
alamandine in mouse isolated aortic rings and in anesthetized brain [42–47]. Protective effects of Ang-(1–7) were also ob-
rats, respectively, were not blocked by the MasR antagonist, served in fructose-fed (FF) rats [48]. In animals subjected to
A-779. In addition, the vasorelaxation effect of alamandine chronic intracerebroventricular infusion, this peptide presented
was not blocked in aortic rings from MasR-knockout mice normalization of mean arterial pressure (MAP) and barorreflex
[11••]. In 2008, by using the Ang-(1–7) antagonist, D-Pro7- bradycardia sensitivity index. In addition, Ang-(1–7) reduced a
Ang-(1–7), Gembardt et al. demonstrated that Ang-(1–7) also cardiac sympathetic tone to lower levels than controls and
binds poorly to Mas-related G protein-coupled type D. Due to fructose-fed rats [48•]. Similar effects were demonstrated in
the high degree of similarity of Ang-(1–7) to alamandine, it was the injection of Ang-(1–7) into the lateral ventricle of the
hypothesized that the latter could also be a MrgD agonist [38•]. DOCA-salt rats’ brain [49]. In this study, central administration
In the first experiment, the vasorelaxation response evoked by of Ang-(1–7) reduced MAP levels, improved barorreflex bra-
alamandine in isolated aortic rings was totally blocked by D- dycardia sensitivity indexes, and reduced also the sympathetic
Pro7-Ang-(1–7). Further testing this hypothesis, the authors ratio/parasympathetic tones [49]. It was also demonstrated by
showed that pre-incubation of arteries with β-alanine, an ago- this same group that transgenic rats that present a lifetime in-
nist of the MrgD receptor, also blocked the effects of crease in circulating levels of Ang-(1–7) and TGR(A1-7)3292
alamandine. Finally, binding assays of alamandine in CHO are protected against exacerbated blood pressure elevation, car-
cells transfected with the MrgD receptor revealed a novel sig- diac dysfunction, and fibrosis induced by DOCA-salt hyperten-
naling pathway for this newly discovered peptide [11••]. MrgD sion [50••].
belongs to a family of G protein-coupled receptors present in The effects of Ang-(1–7) on the autonomic modulation
nociceptors, skeletal muscle, heart, testis, and neurons in ro- were also found by Bertagnolli et al.; they observed an in-
dents, primates, and humans [11••, ]. Since the discovery of crease in heart rate (HR) variability as well as reduction of
alamandine as the new ligand of the MrgD receptor, an increas- low frequency and increase of high frequency in SHR submit-
ing number of studies have been published evidencing the pro- ted to oral administration of Ang-(1–7) included in 2-
tective effects of the activation of this pathway. Up to now, the Hydroxypropyl-β-cyclodextrin (HPβCD) for 10 weeks [51].
specific enzyme that acts to promote descarboxylation of the These authors demonstrated that Ang-(1–7) reduced the diameter
angiotensin is still not known. Despite of that, the presence of of cardiomyocytes in hypertensive animals compared to their
the peptides Ang A and alamandine in the circulation stimulates controls. Corroborating with these results, Ang-(1–7) adminis-
the development of studies in order to establish where, when, tered subcutaneously over a 28-day period was able to prevent
17 Page 4 of 15 Curr Hypertens Rep (2018) 20: 17

Fig. 1 Role of angiotensin-(1–7)


and alamandine in different
organs in experimental
hypertension. The renin-
angiotensin system (RAS) is
critically involved in
pathophysiological changes in
different organs in hypertension.
Among them are the blood
vessels, heart, kidneys, and brain.
Angiotensin-(1–7) [Ang-(1–7)]
and alamandine through the
binding to Mas and Mas-related G
protein-coupled receptor member
D (MrgD) receptors, respectively,
promote protective actions for the
organism, decreasing progression
and attenuating damage caused
by elevated blood pressure,
usually characterized by fibrosis,
remodeling, inflammation, and
excessive vasoconstriction.
CSAR indicates cardiac
sympathetic afferent reflex;
RVLM, rostral ventrolateral
medulla; CVLM, caudal
ventrolateral medulla; RSNA,
renal sympathetic nerve activity

blood pressure rise and reduce sympathetic activity of the renal this experimental model of hypertension, an increase in the
nerve in Sprague-Dawley (SD) rats submitted to intermittent vasodilatory response promoted by Ang-(1–7) in the carotid
chronic hypoxia [52]. In addition, these authors demonstrated artery rings of hypertensive animals was observed, probably
that Ang-(1–7) reduced renal oxidative stress in this model by related to the fact that in these animals Ang-(1–7) and MasR
decreasing levels of malondialdehyde (MDA) and increasing expressions are increased in the carotid artery from 2K1C
superoxide dismutase (SOD) and catalase (CAT) expressions, [56].
significantly attenuating renal fibrosis in these animals. It is noteworthy that the cardioprotective effects can occur
When administered directly in the rostral ventrolateral me- independently of the regulation of blood pressure [57••]. In the
dulla (RVLM) of two-kidney, one clip (2K1C)-operated rats, DOCA-salt hypertension model, Grobe et al. [58] demonstrated
Ang-(1–7) promoted an increase in the renal sympathetic that Ang-(1–7) attenuated cardiac fibrosis without interfering
nerve activity (RSNA) and MAP in 2K1C rats and increased with blood pressure. In this animal model of hypertension, the
cardiac sympathetic afferent reflex and sympathetic output in protective effects of Ang-(1–7) occur through the activation of
renovascular hypertension, effects that were mediated by Ca2+ signaling pathways, reduction of hypertrophic markers, and
MasR [53]. The effect of Ang-(1–7) when microinjected in activation of the Akt signaling pathway, even under conditions of
RVLM was also observed in animals with stress-induced hy- moderately elevated blood pressure [57]. As previously men-
pertension. However, there were no differences in blood pres- tioned, Ang-(1–7) is the major endogenous ligand of MasR.
sure between these animals and their respective controls [54]. Thus, blocking this receptor prevents Ang-(1–7) from promoting
In contrast, microinjection of Ang-(1–7) into the basolateral its protective effects on hypertension. Intracerebroventricular ad-
amygdala (BLA) in Wistar rats attenuated the increase of ministration of A-779 in female SD rats with aldosterone/NaCl-
blood pressure induced by acute emotional stress. This hypo- induced hypertension exacerbated the increase in MAP levels
tensive effect of Ang-(1–7) occurred in a concentration- and thus demonstrating the importance of endogenous central
dependent manner and was blocked by A-779, indicating the Ang-(1–7) in protection against blood pressure elevation [59].
participation of MasR [55]. Subcutaneous administration of the MasR antagonist, A-779, in
Interestingly, in hypertension induced by 2K1C surgery, an Ang II-induced hypertension in SD rats increased recruitment of
exacerbation of the vascular effects of Ang-(1–7) occurred. In macrophages and total collagen deposition in cardiac tissue. In
Curr Hypertens Rep (2018) 20: 17 Page 5 of 15 17

addition, A-779 increased the expression of pro-inflammatory Recently, a new non-peptidic MasR agonist, CGEN-856S,
genes (Gp91, MCP-1, ICAM-1, and caspase 3), and pro-fibrotic has been shown to have effects similar to those promoted by
markers (TGF-β, TIMP-2, and Bcl2). However, in this study Ang-(1–7). In addition to the endothelium-dependent
there were no differences in blood pressure between the group vasorelaxant effect on rat aortic rings, it has antiarrhythmogenic
subjected to Ang II and A-779 co-treatment when compared to effects and promotes reduction in blood pressure in SHR.
the group treated with Ang II alone [60]. A subcutaneous admin- CGEN-856S was also effective in attenuating cardiac remod-
istration of Ang-(1–7) for 4 weeks reduced SBP and diastolic eling in hypertensive rats or experimental model of cardiac
blood pressure (DBP) as well as the Heart Weight/Body Weight hypertrophy [70, 71]. Although it is well established that
(HW/BW) ratio, and reduced cardiomyocyte diameter and vasorelaxant effects of Ang-(1–7) are MasR dependent, recent-
perivascular fibrosis in the heart, an effect that was attributed to ly it was shown that acute bolus infusion of Ang-(1–7) reduces
the activation of the Ang-(1–7)/Sirt3 signaling pathway [61•]. MAP in wild-type mice but not in animals with gene deletion of
Also, in a mouse model of chronic infusion of Ang II over a 4- the MrgD receptor (MrgD Knockout). This result indicates that
week period, Lin et al. demonstrated that Ang-(1–7) also reduced Ang-(1–7) may also bind to the MrgD receptor [72].
cardiac fibrosis and cardiomyocyte diameter, accompanied by an
expressive decrease of the gene expression for atrial natriuretric
peptide (ANP) and skeletal α-actin (SAA), in addition to atten-
Alamandine and Hypertension
uated cardiac oxidative stress via increased SOD activity and
reduced MDA expression [62]. Furthermore, in vitro studies
In the pioneering study of Santos’s group, it was demon-
demonstrated that Ang-(1–7) promotes these cardioprotective
strated that oral administration of alamandine/HPβCD, at a
effects through the activation of the NO/cGMP pathway and
single dose, decreased the blood pressure of SHR. This
inhibition of the prohypertrophic calcineurin/NFAT signaling
was the first demonstration of a long-lasting hypotensive
cascade [63••].
effect of alamandine [11••]. In addition, these authors
Activation of the ACE2/Ang-(1–7)/MasR axis has also
showed that alamandine has pressor and depressor effects
been reported to be protective against the occurrence of stroke
when microinjected into the RVLM and caudal ventrolat-
[64, 65]. Regenhardt et al. demonstrated that central adminis-
eral medulla (CVLM) of Fisher rats, respectively, and these
tration of Ang-(1–7) increased the survival of stroke-prone
effects were blocked by the co-administration of D-Pro7-
spontaneously hypertensive rats (spSHRs) fed a high-salt diet,
Ang-(1–7). Alamandine also had a hypotensive effect
in addition to decreasing the number of subcortical hemor-
when administered to the CVLM of renal hypertensive
rhages [66•]. These authors also demonstrated that the central
rats, 2K1C. In this study, the hypotensive effects of
infusion of Ang-(1–7) improved the neurological status of the
alamandine were blocked by PD123319 and D-Pro7, but
animals and reduced the number of Iba-1-positive cells
not by losartan and A-779 [73].
(microglial marker), without effects on blood pressure and
In a more recent study, Hekmat et al. observed that
cardiac and renal damage parameters that naturally occur in
alamandine presents a biphasic cardiovascular response in
these animals.
2K1C hypertensive rats. [74]. Intravenous administration of
These data were reinforced by recent studies demonstrating
alamandine in animals with renovascular hypertension pro-
the beneficial effects of AVE0991 on blood pressure regula-
duced an AT1R mediated blood pressure pressor effect,
tion and other pathophysiological mechanisms related to car-
followed by a hypotensive response. The fall in blood pressure
diovascular disease. In 2R1C rats, a dose of 3.0 mg/kg of
was mediated, in part, by PD-123319 sensitive receptors.
AVE0991 over a 28-day period reduced MAP, an effect that
Further systematic studies are needed to confirm the binding
was not observed at a dose lower than 1.0 mg/kg. However,
affinity of alamandine to AT1R, AT2R, MasR, and MrgD and
both doses improved baroreflex sensitivity and reduced cardi-
to establish the relative role of the interaction of alamandine
ac fiber thickening [67]. AVE0991 also had mimetic effects on
with these receptors in vivo. Some effects of alamandine and
Ang-(1–7)-induced rat coronary vasodilation, an effect that
Ang-(1–7) on several animal models of hypertension are listed
occurred through the NO-guanylate cyclase pathway.
in Table 1.
Interestingly, this effect was lost in hearts of rats subjected to
coarctation of the abdominal aorta, and restored with the use
of the AT1R antagonist losartan [68]. The effects of AVE0991
on vascular tonus regulation were also observed when chron- Angiotensin-(1–7) and Atherosclerosis
ically administered in SD rats subjected to high salt diet. Oral
treatment with a dose of 10−7 mol/L during 3 days restored the Since the first animal model of atherosclerosis was conceived
acetylcholine-induced vasodilation in the mesenteric arteries and characterized by Anitschkow and Chalatow [75•], many
on high salt diet animals to levels similar to their respective advances in basic science have been made toward a better
controls, without effects on blood pressure [69]. understanding of its pathophysiology [76–78]. The
Table 1 ACE2/Ang-(1–7)/MasR, AVE0991, and alamandine/MrgD in experimental hypertension

Animal model Drug/target Outcomes References

Akita mice (SDA type I) 500 μg/kg/day of Ang-(1–7) administered subcutaneously over a 6-week ↓ SBP [40]
17 Page 6 of 15

period
(mRen2)27 ICV infusion of 200 ng/h of Ang-(1–7) ↓ SBP and DBP [41]
↓ Cardiomyocyte hypertrophy
↓ Collagen deposition
↓ Type I collagen
↓ Fibronectin
SHR Oral administration of 72 μg/kg−1/day−1 of Ang-(1–7)/HP-βCD Improve HR variability [51]
↓ Reduction of low frequency and increase of high frequency
↓ Diameter of cardiomyocytes
SD rats—intermittent chronic hypoxia 400 ng/kg−1/min−1 of Ang-(1–7) administered subcutaneously over a 28-day Prevent blood pressure rise [52]
period ↓ RSNA
↓ MDA
↑ SOD
↑ CAT
2K1C rats RVLM microinjection of Ang-(1–7) (0.3 nmol) ↑ RSNA [53]
↑ MAP
↑ CSAR
↑ Sympathetic output
SIH rats RVLM microinjection of Ang-(1–7) (100 pmol) ↑ MAP [54]
No differences between groups
2K1C rats Cumulative concentration-response curves ↑ Vasodilatory response promoted by Ang-(1–7) in the carotid artery [56]
Ang-(1–7)—0.1 nmol/L–1 μmol/L rings
Female SD rats—aldosterone/NaCl-induced A-779 ICV infusion ↑ Increase in MAP levels [59]
hypertension 60 ng kg−1 min−1
Ang II-induced hypertension in SD rats A-779 at 1 mg/day administered subcutaneously for 4 weeks ↑ Recruitment of macrophages [60]
↑ Total collagen deposition in cardiac tissue
↑ Gp91, MCP-1, ICAM-1, and caspase 3
↑ TGF-β, TIMP-2, and Bcl2
No differences on blood pressure
Ang II-induced hypertension in SD rats 400 ng/kg/min of Ang-(1–7) administered subcutaneously for 4 weeks ↓ SBP and DBP [61•]
↓ HW/BW ratio
↓ Reduced cardiomyocyte diameter
↓ Perivascular fibrosis in the heart
C57BL/6 mice and chronic infusion of Ang II Subcutaneous administration of 500 μg/kg/day of Ang-(1–7) for 4 weeks ↓ Cardiac fibrosis and cardiomyocyte diameter [62]
↓ ANP ↓ SAA
↑ SOD activity
↓ MDA expression
spSHRs ICV infusion of Ang-(1–7)—1.0 μg in 0.15 μL ↑ The survival [66•]
ACSF/h ↓ The number of subcortical hemorrhages
Improved the neurological status
↓ The number of Iba-1-positive cells
FF rats Chronic intracerebroventricular infusion of 200 ng/h of Ang-(1–7) for Normalization of MAP and barorreflex bradycardia sensitivity index [48•]
4 weeks ↓ Cardiac sympathetic tone and
↓ Sympathetic ratio/parasympathetic tones
DOCA-salt rats Chronic intracerebroventricular infusion of 200 ng/h of Ang-(1–7) for ↓ MAP [49]
17 days ↑ Baroreflex bradycardia sensitivity index
Curr Hypertens Rep (2018) 20: 17
Curr Hypertens Rep (2018) 20: 17 Page 7 of 15 17

2K1C two-kidney, one clip, (mRen2)27 transgenic hypertensive rats, CAT catalase, CSAR cardiac sympathetic afferent reflex, DBP diastolic blood pressure, HP-βCD HP-β cyclodextrin, HR heart rate, ICV
intracerebroventricular, MAP mean arterial pressure, MDA malondialdehyde, SBP systolic blood pressure, SD Sprague-Dawley, SDA spontaneously diabetic animals, SHR spontaneously hypertensive rats,
References

RSNA renal sympathetic nerve activity, SOD superoxide dismutase, RVLM rostral ventrolateral medulla, SIH stress-induced hypertension, spSHRs stroke-prone spontaneously hypertensive rats, FF
cardiovascular effects evoked by the RAS depend on the bal-
ance between the classical and protective arms of RAS in the

[11••]
[70]

[74]
[73]
[69]
[67]
vessel layers [79]. Thus, among other risk factors for athero-
sclerosis, RAS dysregulation and/or overactivity has been im-

Restored the acetylcholine-induced vasodilation in the mesenteric


plicated as one of the leading mechanisms in atherosclerosis,
participating in all phases of the disease, from plaque devel-
opment and progression until its rupture [80].

Biphasic reponses on blood pressure receptor mediated


Within the cardiovascular system, most of the RAS com-
ponents have been previously reported to be present in the
heart and blood vessels, including veins, aorta, and small re-
Endothelium-dependent vasorelaxant effect
↓ Sympathetic ratio/parasympathetic tones

sistance arteries [79]. Evidence of the existence of a vascular


tissue RAS includes the generation and expression of compo-
nents of both classical and protective arms in blood vessels,
↑ Baroreflex sensitivity and

such as renin, angiotensinogen, ACE, Ang I, Ang II, AT1R,


Antiarrhythmogenic effect
↓ Cardiac fiber thickening

AT2R, ACE2, Ang-(1–7), MasR, alamandine, and MrgD re-


ceptor [11••, 81–84]. In this topic, we focus on the importance
of the protective arms of RAS for atheroprotection and atten-
uation of atherosclerotic lesions in animal models and obser-
Outcomes

arteries

↓ MAP
↓ MAP

↓ MAP
↓ MAP

vations in humans, which may provide an insight into future


therapeutic approaches.
Zulli et al. [85] first identified both ACE2 and AT2R ex-
Administration of AVE0991 (10–7 M) during 3 days in the drinking water

pressions in macrophages and VSMC within the atheroscle-


rotic plaque in a New Zealand White (NZW) rabbit model of
Oral administration of 3.0 mg/kg of AVE0991 over a 28-day period

Intravenous administration of a single 75 μg/kg dose of alamandine

atherosclerosis. In fact, Tesanovic et al. [86••] noticed for the


first time, in atherosclerotic lesions of ApoE knockout (ApoE-
Oral administration of alamandine/HPβCD at a single dose

KO) mice fed a high-fat diet, that long-term Ang-(1–7) treat-


ment alone diminished the longitudinal development of the
CVLM microinjection of alamandine (4, 40 pmol)

lesions, which was totally and partially reversed by A-779


Acute infusion of graded doses of CGEN-856S

and PD-123319. As by that time, PD-123319 was considered


Cumulative concentration-response curves

as a selective AT2R antagonist, the authors discussed the po-


CGEN-856S—0.0001 to 1 μmol/L and

t e n t i a l c r o s s t a l k b e t w e e n M a s R a n d AT 2 R f o r
atheroprotection and vasoprotection [86••]. More recent stud-
ies have identified PD-123319 also as an MrgD receptor an-
tagonist; thus, we cannot exclude the possibility that
alamandine is formed from Ang-(1–7) in this model, and pro-
tective effects observed are partially mediated by alamandine/
fructose-fed rats, DOCA-salt deoxycorticosterone acetate salt rats

MrgD signaling activation [11••].


Drug/target

Dong et al. [87], in a similar rabbit model of Zulli et al. [85],


with a gene transfer-based strategy, injected a one-time infusion
of transfected plasmids for ACE2 (AdACE2), showing a reduc-
tion in the intimal vessel layer thickness and the intimal-to-
medial ratio. Additionally, they showed an alteration of angio-
tensin metabolism toward an increased ACE2 expression and
activity, suggesting that the manipulation of its expression may
delay and stabilize the atherosclerotic process [87]. These au-
thors also reported that the intimal macrophage content and
SD rats—high salt (HS) diet

MMP-3 and MMP-9 expression and activity were diminished


and, despite unaltered VSMC numbers, there was increased
Table 1 (continued)

collagen production, which altogether contributed to the stabi-


Animal model

lization of atherosclerotic lesions [87]. Furthermore, the


AdACE2 treatment approach conducted in ApoE-KO mice
2K1C rats
2K1C rats
2K1C rats

fed Western-type diet for a month lowered serum triglyceride,


SHR
SHR

lipid content, and monocyte/macrophage accumulation within


17 Page 8 of 15 Curr Hypertens Rep (2018) 20: 17

the atherosclerotic plaque [88, 89]. In 2008, a study involving a proatherogenic MMP, through suppression of p38 MAPK
elder human patients, most of them men at different stages of activity but stated that lower collagen I production in VSMC
atherosclerosis, was performed by an in situ binding tech- from ApoE-KO mice fed a high-fat diet helped in stabilizing
nique, which indicated that ACE isoforms were expressed atherosclerotic plaques. [96]. Altogether, these results showed
at both transcript and protein levels in diseased and non- the importance of the MasR in the negative regulation of the
diseased vessels [90], showing a similar pattern of expression mechanisms involved in the atherogenic process and the in-
throughout the atherosclerosis process. ACE2 activity, how- fluence of the diet type in the facilitation of atherosclerosis.
ever, was significantly diminished from early to stable lesion Through the years, some Ang-(1–7) analogues have been
and increased from stable to thrombus-containing lesions, synthesized and used in experimental protocols, such as
suggesting that the protective action of the contra-regulatory AVE0991, CGEN-856S, and others [97]. In the literature, only
axis of the RAS is somehow lost in that process. Using the AVE0991 has been used to mimic Ang-(1–7) actions on ath-
previously described rabbit in vivo model [87] in association erosclerotic models [98–100]. Using ApoE-KO mice fed with
with in vitro HUVEC and VSMC cell cultures, Zhang et al. chow diet in combination with AVE0991 treatment for
[91] confirmed previous data obtained with ACE2 expression 4 months, Toton-Zuranska et al. [98] reported that no changes
by AdACE2 infusion in plaques [85], showing a reduction in in cholesterol and triglycerides were observed in the plasma of
morphological parameters, and showed its occurrence in vitro these animals, but AVE0991 was able to reduce the athero-
[87]. AdACE2 treatment also increased Ang-(1–7) expres- sclerotic lesion area in both en face and cross-section methods
sion both in vivo and in vitro while abrogating ACE and of aortic roots. Regarding the evaluation of the plasma levels
AT1R expressions. Furthermore, AdACE2 decreased the ex- of important pro-inflammatory indicators such as IL-6, MCP-
pression of inflammatory mediators, such as MCP-1 in vitro, 1, IL-12, and serum amyloid A (SaA), Jawien et al. [99] re-
and suppressed the growth of VSMC and lowered macro- ported that the treatment of ApoE-KO mice with AVE0991
phage accumulation in neointima [91]. was able to lower them, besides the area of lipid and athero-
In a study from our group, using the mouse model of shear sclerotic changes in the aorta and in the aortic root, and VSMC
stress-induced atherogenesis and plaque vulnerability in and macrophage staining as compared to the control group
ApoE-KO mice, we reported that a 3-week treatment with ApoE-KO mice after 4 months of treatment [101]. In the same
Ang-(1–7)/HPβCD downregulates collagen, lipid, and im- animal model, Olszanecki et al. [100] assessed Ang-(1–7) and
mune cell contents in atherosclerotic plaques; however, dis- Ang II concentrations through Ang I metabolism by mass
tinct shear stress-prone regions respond differently to the treat- spectrometry in an ex vivo experimental protocol, which dem-
ment [92••]. In a closer examination of the inflammation pro- onstrated that ApoE-KO mice naturally express higher levels
cess within the atherosclerotic plaque, Silva et al. [93], using of Ang II, but not Ang-(1–7). It was shown that the increased
standard diet-fed MasR/ApoE-double-knockout (DKO) mice, synthesis of Ang II in atherosclerotic thoracic aorta from
observed that decreased fat mass and worsened dyslipidemia ApoE-KO mice was significantly reduced by treatment with
resulted in an increased pro-atherogenic environment without AVE0991, but no differences in Ang-(1–7) production were
affecting the size of aortic root lesions, despite the reduced observed with or without diet. Another insight into the role of
leptin serum levels. The lipid metabolic alterations observed Ang-(1–7) in atherosclerotic plaques by orally administered
in the DKO mice were partially inferred as one of the causes AVE0991 was provided by Skiba et al. [102•]. In female
for the absence of increased atherogenesis and the unaltered ApoE-KO mice, AVE0991 treatment was able to reduce the
intraplaque contents of leukocytes, VSMC and MMPs, except pro-inflammatory responses of macrophages and peripheral
for the macrophage accumulation, which was reduced [93]. blood monocytes, as well as macrophage and leukocyte infil-
However, using Mas/ApoE-DKO mice fed a high-fat Western tration within the plaques by inhibiting the production of pro-
diet for 3 months, it was observed that the aortic lesion area in inflammatory cytokines in the perivascular adipose tissue,
DKO mice was bigger and presented more accumulation of which also has a similar MasR expression as to the control
elicited mononuclear cells, which expressed higher levels of group. As previously reported, the atherosclerotic lesion area
pro-inflammatory interleukins than in ApoE-KO mice [94•]. in the aortic arch and thoracic aorta decreased [102•]. Figure 2
The deficiency in the MasR in those animals also affected shows some of the effects mediated by protective arms of
macrophage gene expression, inducing a pro-inflammatory RAS in experimental atherosclerosis.
phenotype in the aorta [94•]. Besides reporting the positive
effects of a 4-week Ang-(1–7) treatment on the size of athero-
sclerotic lesions, Yang et al. [95] observed an increase in col-
lagen and VSMC content combined with lower numbers of Alamandine and Atherosclerosis
inflammatory macrophages and pro-inflammatory cytokines
in ApoE-KO mice. In another study, it was reported that a The recent discovery and characterization of alamandine and
long-term Ang-(1–7) treatment was able to reduce MMP-8, its receptor MrgD added a new level of complexity to the
Curr Hypertens Rep (2018) 20: 17 Page 9 of 15 17

Fig. 2 The effects of angiotensin-(1–7), AVE-0991, and alamandine on stabilizing atherosclerotic plaques and in regulating some blood and
the pathophysiology of atherosclerosis. Within the vascular wall, there is intraplaque atherosclerotic markers (right panel). The activation of the
a highly inflammatory and necrotic microenvironment, which contributes protective arm of RAS by Ang-(1–7) and AVE-0991 through the Mas
to immune cell recruitment and migration, as well as increased lipid receptor (MasR) reduces plaque morphological parameters and
accu mulation, low-density protein (LDL) oxidation, and inflammatory cytokine production, while increasing collagen and
metalloproteinase (MMPs) activity. Altogether, these events induce angiotensin-converting enzyme 2 (ACE2) and Ang-(1–7) expressions
atherosclerosis, in which different cell types are involved, such as (in blue). Alamandine, a newly characterized RAS peptide, has shown
macrophages, endothelial cells, and vascular smooth muscle cells to have some similar effects as to Ang-(1–7) through Mas-related G
(VSMC) (left panel). Animal models have provided useful information protein-coupled receptor member D (MrgD), but reports on the
on how atherosclerosis arises and advances; and usage of renin- induction of plasminogen activator inhibitor-1 (PAI-1) highlights the
angiotensin system (RAS) peptides or analogues, such as angiotensin- demand that further investigation of this peptide role in atherosclerosis
(1–7) [Ang-(1–7)], AVE0991, and alamandine, has been helpful in is conducted (in black)

known RAS [11••]. Lautner, Vilella, Fraga-Silva et al. [11••] intraplaque collagen deposition, which suggests no ameliora-
showed that alamandine evokes similar central cardiovascular, tion of plaque structural stability [104••]. Uchiyama et al.
antihypertensive, and vasodilatory effects as to those by [105•], using Wistar rats fed a chow diet, showed that a 2-
Ang-(1–7). To date, there are only three reports on the effects day treatment with alamandine or Ang-(1–7) had opposing
of alamandine in atherosclerotic plaques [103–105•]. effects on leptin expression and secretion dynamics.
Habiyakare et al. [103] first reported the presence of the Alamandine, through the MrgD, receptor also evoked con-
MrgD receptor in healthy and diseased aortae and its co- flicting effects over two pro-atherogenic proteins in vitro and
localization with eNOS-positive cells and, possibly, in macro- in vivo such as reduction of leptin expression and secretion in
phages in the NZW rabbit model fed a 1-month high-fat diet. adipocytes, but increased plasminogen activator inhibitor-1
They also observed that the vasodilatory effects of alamandine (PAI-1) [105•]. It is possible that alamandine may act more
were reduced in thoracic, carotid, renal, and iliac arteries [103]. as a protector of the endothelial function than as a plaque
Da Silva et al., using a cast device in ApoE-KO mice fed stabilizer.
Western-type diet, demonstrated that alamandine treatment The reports mentioned above have helped to identify the
for 1 month reduced degranulation of neutrophils in vitro and dynamics of the newly characterized alamandine/MrgD path-
in vivo, besides decreasing the levels of circulating and way in atherosclerosis, but further research is needed to better
intraplaque MMP-9 [104••]. However, no changes in lipid, address its role in the atherosclerotic process. Table 2 summa-
macrophage, and neutrophil contents or the MrgD expression rizes the effects of Ang-(1–7), AVE0991, and alamandine on
at both mRNA and protein levels were observed, as well as experimental atherosclerosis.
Table 2 ACE2/Ang-(1–7)/MasR, AVE0991, and alamandine/MrgD in experimental atherosclerosis

Animal model Drug/target Outcomes References

NZW rabbit One-time infusion of AdACE2 suspension (2.5 × 109 pfu/mL) ↓ Intimal vessel layer thickness and intimal-to-medial ratio [87, 91]
↓ Growth of VSMC and neointimal macrophage accumulation
17 Page 10 of 15

↓MMP-3 and MMP-9 expression


↓ ACE and AT1R expressions
↑ ACE2 activity and collagen production
↑ ACE2 and Ang-(1–7) expressions

ApoE-KO mice One-time infusion of AdACE2 suspension (2.5 × 109 pfu/mL) ↓ Lipid and monocyte/macrophage accumulation [88, 89]
↓ Serum levels of MCP-1, VCAM-1 and IL-6
↑ Neovascularization

ApoE-KO mice Subcutaneously administered Ang-(1–7) (24 or 48 g/kg per hour, 1 month) ↓ Longitudinal development of the lesions [86••]

ApoE-KO mice Ang-(1–7) with or without other drugsVarious doses (100–400 ng/kg/min, ↑ Collagen and VSMC content [95]
1 month) ↓ Size of atherosclerotic lesion, numbers of inflammatory macrophages, and
pro-inflammatory cytokines

ApoE-KO mice Ang-(1–7) with or without other drugs (400 ng/kg/min, 1 month) ↓ MMP-8 activity and collagen I production in VSMC [96]

ApoE-KO mice + Ang-(1–7)/HPβCD by gavage (≈ 30 μg/kg/day of Ang-(1–7) for 5 days/week, ↓ Collagen, lipid, and immune cell contents [92••]
cast 3 weeks)
MasR/ApoE-DKO MasR deficiency ↓ Leptin serum levels and macrophage accumulation [93, 94•]
mice ↑ Size of aortic root lesions
↑ Aortic lesion area in DKO mice
↑ Accumulation of immune cells and levels of inflammatory interleukins
Absence of increased atherogenesis
Wistar rats Injection of Ang-(1–7)Various doses, 2 × 2 days, interval 24 h ↑ Leptin expression and secretion in adipocytes [105•]

ApoE-KO mice AVE0991 combined diet (0.58 μmol/kg b.w./day, 4 months) ↓ Pro-inflammatory cytokines in plasma [98–101]
↓ VSMC and macrophage staining
↓ Atherosclerotic lesion area
↓ Area of lipid changes in aorta
↓ Area of atherosclerotic changes
↓ Increased tissue synthesis of Ang II
No differences in tissue Ang-(1–7) levels
No change in cholesterol and triglycerides in blood

ApoE-KO mice AVE0991 combined diet (0.58 μmol/kg b.w./day, 1–3 months) ↓ Inflammatory response of macrophages and peripheral blood monocytes [102]
↓ Immune cells infiltration
↓ Production of pro-inflammatory cytokines in pVAT
↓ Atherosclerotic lesion area

ApoE-KO mice Subcutaneously administered alamandine (24 μg/kg/h, 1 month) ↓ Degranulation of neutrophils and levels of circulating and intraplaque MMP-9 [104••]
No changes in intraplaque collagen, lipid, macrophage, and neutrophil contents or
the MrgD expression

Wistar rats Injection of alamandineVarious doses (2 days, interval 24 h) ↓ Leptin expression and secretion in adipocytes [105•]
↑ PAI-1 expression in adipocytes
Curr Hypertens Rep (2018) 20: 17
Curr Hypertens Rep (2018) 20: 17 Page 11 of 15 17

Compliance with Ethical Standards References

Conflict of Interest The authors declare no conflicts of interest relevant Papers of particular interest, published recently, have been
to this manuscript. All the authors have declared that there is nothing to highlighted as:
disclosure. • Of importance
•• Of major importance
Human and Animal Rights and Informed Consent This article does not
contain any studies with human or animal subjects performed by any of
1. (2015) Global, regional, and national age-sex specific all-cause
the authors.
and cause-specific mortality for 240 causes of death,
1990–2013: A systematic analysis for the global burden of disease
Abbreviations and Acronyms 2K1C, Two-kidney, one clip; study 2013. Lancet 385(9963):117–71. https://doi.org/10.1016/
Ang-(1–7), Angiotensin-(1–7); A-779, D-Ala7- S0140-6736(14)61682-2
2. Granger DN, et al. Microvascular responses to cardiovascular risk
Angiotensin-(1–7); ACE, Angiotensin-converting enzyme;
factors. Microcirculation. 2010;17(3):192–205. https://doi.org/10.
ACE2, Angiotensin-converting enzyme 2; AdACE2, 1111/j.1549-8719.2009.00015.x.
Transfected plasmids for ACE2; Ang A, Angiotensin A; Ang I, 3. Tunon J, et al. Common pathways of hypercholesterolemia and
Angiotensin I; Ang II, Angiotensin II; Ang III, hypertension leading to atherothrombosis: the need for a global
Angiotensin-(2–8); Ang IV, Angiotensin-(3–8); Ang-(1–4), approach in the management of cardiovascular risk factors. Vasc
Health Risk Manag. 2007;3(4):521–6.
Angiotensin-(1–4); Ang-(1–5), Angiotensin-(1–5); Ang-(1–9), 4. Bader M. ACE2, angiotensin-(1-7), and Mas: the other side of the
Angiotensin-(1–9); ANP, Atrial natriuretric peptide; ApoE- coin. Pflugers Arch. 2013;465(1):79–85. https://doi.org/10.1007/
KO, ApoE knockout; AT1R, Angiotensin receptor type I; s00424-012-1120-0.
AT2R, Angiotensin receptor type II; AVP, arginine vasopres- 5. Bader M, Peters J, Baltatu O, Müller DN, Luft FC, Ganten D.
sin; Bcl2, B-cell lymphoma 2; BLA, basolateral amygdala; Tissue renin-angiotensin systems: new insights from experimental
animal models in hypertension research. J Mol Med (Berl).
BW, body weight; CAT, Catalase; CHO cells, Chinese hamster 2001;79(2–3):76–102. https://doi.org/10.1007/s001090100210.
ovary cells; CVD, cardiovascular diseases; CVLM, caudal 6. Fraga-Silva RA, Ferreira AJ, Dos Santos RA. Opportunities for
ventrolateral medulla; DBP, Diastolic blood pressure; DKO, targeting the angiotensin-converting enzyme 2/angiotensin-(1-7)/
Double-knockout; DOCA-salt, Deoxycorticosterone acetate mas receptor pathway in hypertension. Curr Hypertens Rep.
2013;15(1):31–8. https://doi.org/10.1007/s11906-012-0324-1.
salt; D-Pro7Ang-(1-7), D-Pro7Angiotensin-(1-7); GMP,
7. Zaman MA, Oparil S, Calhoun DA. Drugs targeting the renin-
Guanosine monophosphate; Gp91, subunit of phagocyte angiotensin-aldosterone system. Nat Rev Drug Discov.
NADPH oxidase; HF, High frequency; HNS, hypothalamo- 2002;1(8):621–36. https://doi.org/10.1038/nrd873.
neurohypophysial system; HPβCD, 2-Hydroxypropyl-β-cy- 8. Mazzolai L, Hayoz D. The renin-angiotensin system and athero-
clodextrin; HR, Heart rate; HUVEC, Human umbilical vein sclerosis. Curr Hypertens Rep. 2006;8(1):47–53. https://doi.org/
10.1007/s11906-006-0040-9.
endothelial cells; HW, heart weight; ICAM-1, Intercellular
9. Durante A, Peretto G, Laricchia A, Ancona F, Spartera M,
adhesion molecule-1; IL-6, Interleukin-6; IL-12, Interleukin- Mangieri A, et al. Role of the renin-angiotensin-aldosterone sys-
12; MAP, Mean arterial pressure; MasR, Mas receptor; MCP- tem in the pathogenesis of atherosclerosis. Curr Pharm Des.
1, Monocyte chemoattractant protein-1; MDA, 2012;18(7):981–1004. https://doi.org/10.2174/
138161212799436467.
Malondialdehyde; MMPs, Matrix metalloproteinases; MMP-
10. Husain K, Hernandez W, Ansari RA, Ferder L. Inflammation,
3, Matrix metalloproteinases 3; MMP-8, Matrix metallopro- oxidative stress and renin angiotensin system in atherosclerosis.
teinases 8; MMP-9, Matrix metalloproteinases 9; (mRen2)27, World J Biol Chem. 2015;6(3):209–17. https://doi.org/10.4331/
Hypertensive transgenic rats; MrgD, Mas-related G protein- wjbc.v6.i3.209.
coupled type D; NFAT, Nuclear factor of activated T-cells; NO, 11.•• Lautner RQ, Villela DC, Fraga-Silva RA, Silva N, Verano-Braga
T, Costa-Fraga F, et al. Discovery and characterization of
Nitric oxide; NZW, New Zealand white; p38 MAPK, p38
alamandine: a novel component of the renin-angiotensin system.
mitogen-activated protein kinases; PAI-1, Plasminogen acti- Circ Res. 2013;112(8):1104–1111. This paper reports the dis-
vator inhibitor-1; RAS, Renin-angiotensin system; ROS, covery and characterization of alamandine, providing first
Reactive oxygen species; RSNA, Renal sympathetic nerve experimental data on its biological activity, biochemical profile
and interaction with its receptor, MrgD. https://doi.org/10.
activity; RVLM, Rostral ventrolateral medulla; SAA,
1161/CIRCRESAHA.113.301077.
Skeletal α-actin; SaA, Serum amyloid A; SBP, Systolic 12. Dickson ME, Sigmund CD. Genetic basis of hypertension:
blood pressure; SD, Sprague-Dawley; SHR, Spontaneous revisiting angiotensinogen. Hypertension. 2006;48(1):14–20.
hypertensive rats; SOD, Superoxide dismutase; TGF-1, https://doi.org/10.1161/01.HYP.0000227932.13687.60.
Transforming growth factor-1; TIMP-2, Tissue inhibitor of 13. Patel S, Rauf A, Khan H, Abu-Izneid T. Renin-angiotensin-
aldosterone (RAAS): the ubiquitous system for homeostasis and
metalloproteinases-2; VCAM-1, vascular cell adhesion pathologies. Biomed Pharmacother. 2017;94:317–25. https://doi.
protein-1; VSMC, Vascular smooth muscle cells org/10.1016/j.biopha.2017.07.091.
17 Page 12 of 15 Curr Hypertens Rep (2018) 20: 17

14. Bader M, Ganten D. Regulation of renin: new evidence from 23. Using radioimmunoassays and high-performance liquid
cultured cells and genetically modified mice. J Mol Med (Berl). chromatography, the authors described for first time Ang-
2000;78(3):130–9. https://doi.org/10.1007/s001090000089. (1–7) as an endogenous product of the renin-angiotensin sys-
15. Gonzalez AA, Prieto MC. Roles of collecting duct renin and tem, and detected in brain, adrenal and rat plasma.
(pro)renin receptor in hypertension: mini review. Ther Adv 31. Chappell MC. Biochemical evaluation of the renin-angiotensin
Cardiovasc Dis. 2015;9(4):191–200. https://doi.org/10.1177/ system: the good, bad, and absolute? Am J Physiol Heart Circ
1753944715574817. Physiol. 2016;310(2):H137–52. https://doi.org/10.1152/ajpheart.
16. Gonzalez AA, et al. Angiotensin II stimulates renin in inner med- 00618.2015.
ullary collecting duct cells via protein kinase C and independent of 32. Ferrario CM, et al. Advances in the renin angiotensin system focus
epithelial sodium channel and mineralocorticoid receptor activity. on angiotensin-converting enzyme 2 and angiotensin-(1-7). Adv
Hypertens. 2011;57(3):594–99. Pharmacol. 2010;59:197–233. https://doi.org/10.1016/S1054-
17. Lavoie JL, Liu X, Bianco RA, Beltz TG, Johnson AK, Sigmund 3589(10)59007-0.
CD. Evidence supporting a functional role for intracellular renin in 33.•• Santos RA, et al. Angiotensin-(1–7) is an endogenous ligand for
the brain. Hypertension. 2006;47(3):461–6. https://doi.org/10. the G protein-coupled receptor Mas. Proc Natl Acad Sci U S A.
1161/01.HYP.0000203308.52919.dc. 2003;100(14):8258–8263. By using transgenic mice and
18. Nguyen G. Renin/prorenin receptors. Kidney Int. 2006;69(9): in vitro transfected cell model, the authors demonstrated for
1503–6. https://doi.org/10.1038/sj.ki.5000265. the first time the interaction between Ang-(1–7) and the Mas
19. Villela DC, Passos-Silva DG, Santos RA. Alamandine: a new receptor. https://doi.org/10.1073/pnas.1432869100.
member of the angiotensin family. Curr Opin Nephrol 34. Santos RA, Ferreira AJ, Simoes ESAC. Recent advances in the
Hypertens. 2014;23(2):130–4. https://doi.org/10.1097/01.mnh. angiotensin-converting enzyme 2-angiotensin(1-7)-Mas axis. Exp
0000441052.44406.92. Physiol. 2008;93(5):519–27. https://doi.org/10.1113/expphysiol.
20. Bernstein KE, Gonzalez-Villalobos RA, Giani JF, Shah K, 2008.042002.
Bernstein E, Janjulia T, et al. Angiotensin-converting enzyme 35. McCollum LT, Gallagher PE, Ann Tallant E. Angiotensin-(1–7)
overexpression in myelocytes enhances the immune response. attenuatesangiotensin II-induced cardiac remodeling associated
Biol Chem. 2014;395(10):1173–8. https://doi.org/10.1515/hsz- with upregulation of dual-specificityphosphatase 1. Am J
2013-0295. Physiol Heart Circ Physiol. 2012;302(3):H801–H810.
21. Crisan D, Carr J. Angiotensin I-converting enzyme: genotype and 36. Santos, RA. Angiotensin-(1–7). Hypertens. 2014;63(6):1138–47.
disease associations. J Mol Diagn. 2000;2(3):105–15. https://doi. 37. Jankowski V, Vanholder R, van der Giet M, Tölle M, Karadogan
org/10.1016/S1525-1578(10)60624-1. S, Gobom J, et al. Mass-spectrometric identification of a novel
22. Teixeira LG, et al. Conformational properties of seven Toac- angiotensin peptide in human plasma. Arterioscler Thromb Vasc
labeled angiotensin I analogues correlate with their muscle con- Biol. 2007;27(2):297–302. https://doi.org/10.1161/01.ATV.
traction activity and their ability to act as ACE substrates. PLoS 0000253889.09765.5f.
One. 2015;10(8):e0136608. https://doi.org/10.1371/journal.pone. 38.• Gembardt F, Grajewski S, Vahl M, Schultheiss HP, Walther T.
0136608. Angiotensin metabolites can stimulate receptors of the Mas-
23. Ahmad S, Simmons T, Varagic J, Moniwa N, Chappell MC, related genes family. Mol Cell Biochem. 2008;319(1–2):115–
Ferrario CM. Chymase-dependent generation of angiotensin II 123. This article summarizes the findings of the new compo-
from angiotensin-(1-12) in human atrial tissue. PLoS One. nents of the renin-angiotensin system with homologous se-
2011;6(12):e28501. https://doi.org/10.1371/journal.pone. quences to Ang-(1–7) and how they interact with the Mas
0028501. receptor. https://doi.org/10.1007/s11010-008-9884-4.
24. Biancardi VC, Bomfim GF, Reis WL, al-Gassimi S, Nunes KP. 39. van Twist DJ, Kroon AA, de Leeuw PW. Angiotensin-(1-7) as a
The interplay between angiotensin II, TLR4 and hypertension. strategy in the treatment of hypertension? Curr Opin Nephrol
Pharmacol Res. 2017;120:88–96. https://doi.org/10.1016/j.phrs. Hypertens. 2014;23(5):480–6. https://doi.org/10.1097/MNH.
2017.03.017. 0000000000000050.
25. Lacolley P, Safar ME, Regnault V, Frohlich ED. Angiotensin II, 40. Shi Y, Lo CS, Padda R, Abdo S, Chenier I, Filep JG, et al.
mechanotransduction, and pulsatile arterial hemodynamics in hy- Angiotensin-(1-7) prevents systemic hypertension, attenuates ox-
pertension. Am J Physiol Heart Circ Physiol. 2009;297(5): idative stress and tubulointerstitial fibrosis, and normalizes renal
H1567–75. https://doi.org/10.1152/ajpheart.00622.2009. angiotensin-converting enzyme 2 and Mas receptor expression in
26. Weiss D, Sorescu D, Taylor WR. Angiotensin II and atheroscle- diabetic mice. Clin Sci (Lond). 2015;128(10):649–63. https://doi.
rosis. Am J Cardiol. 2001;87(8A):25C–32C. org/10.1042/CS20140329.
27.•• Santos RA, Brosnihan KB, Chappell MC, Pesquero J, Chernicky 41. Kangussu LM, Guimaraes PS, Nadu AP, Melo MB, Santos RAS,
CL, Greene LJ, et al. Converting enzyme activity and angiotensin Campagnole-Santos MJ. Activation of angiotensin-(1-7)/Mas axis
metabolism in the dog brainstem. Hypertension. 1988;11(2 Pt 2): in the brain lowers blood pressure and attenuates cardiac remod-
I153–I157. This paper reports for the fist time the presence of eling in hypertensive transgenic (mRen2)27 rats.
Ang-(1–7) and ECA activity in the dog brain. https://doi.org/10. Neuropharmacology. 2015;97:58–66. https://doi.org/10.1016/j.
1161/01.HYP.11.2_Pt_2.I153. neuropharm.2015.04.036.
28. Schiavone MT, Santos RA, Brosnihan KB, Khosla MC, Ferrario 42. Diez-Freire C, et al. ACE2 gene transfer attenuates hypertension-
CM. Release of vasopressin from the rat hypothalamo- linked pathophysiological changes in the SHR. Physiol Genomics.
neurohypophysial system by angiotensin-(1-7) heptapeptide. 2006;27(1):12–9. https://doi.org/10.1152/physiolgenomics.
Proc Natl Acad Sci U S A. 1988;85(11):4095–8. https://doi.org/ 00312.2005.
10.1073/pnas.85.11.4095. 43. Feng Y, Xia H, Cai Y, Halabi CM, Becker LK, Santos RAS, et al.
29. Campagnole-Santos MJ, et al. Cardiovascular effects of angioten- Brain-selective overexpression of human angiotensin-converting
sin-(1-7) injected into the dorsal medulla of rats. Am J Phys. enzyme type 2 attenuates neurogenic hypertension. Circ Res.
1989;257(1 Pt 2):H324–9. 2010;106(2):373–82. https://doi.org/10.1161/CIRCRESAHA.
30.•• Chappell MC, Brosnihan KB, Diz DI, Ferrario CM. Identification 109.208645.
of angiotensin-(1–7) in rat brain. Evidence for differential process- 44. Feng Y, Yue X, Xia H, Bindom SM, Hickman PJ, Filipeanu CM,
ing of angiotensin peptides. J Biol Chem. 1989;264(28):16518– et al. Angiotensin-converting enzyme 2 overexpression in the
Curr Hypertens Rep (2018) 20: 17 Page 13 of 15 17

subfornical organ prevents the angiotensin II-mediated pressor relaxation response in renovascular hypertensive rat carotid.
and drinking responses and is associated with angiotensin II type Peptides. 2015;71:250–8. https://doi.org/10.1016/j.peptides.
1 receptor downregulation. Circ Res. 2008;102(6):729–36. https:// 2015.08.002.
doi.org/10.1161/CIRCRESAHA.107.169110. 57.•• de Almeida PW, et al. Beneficial effects of angiotensin-(1–7)
45. Sriramula S, Cardinale JP, Lazartigues E, Francis J. ACE2 over- against deoxycorticosterone acetate-induced diastolic dysfunction
expression in the paraventricular nucleus attenuates angiotensin II- occur independently of changes in blood pressure. Hypertension.
induced hypertension. Cardiovasc Res. 2011;92(3):401–8. https:// 2015;66(2):389–395. In this work the authors demonstrated
doi.org/10.1093/cvr/cvr242. that the signaling pathways involved in the cardioprotective
46. Xia H, Sriramula S, Chhabra KH, Lazartigues E. Brain effects of Ang-(1–7) are activated even under conditions of
angiotensin-converting enzyme type 2 shedding contributes to high blood pressure. https://doi.org/10.1161/
the development of neurogenic hypertension. Circ Res. HYPERTENSIONAHA.114.04893.
2013;113(9):1087–96. https://doi.org/10.1161/CIRCRESAHA. 58. Grobe JL, Mecca AP, Mao H, Katovich MJ. Chronic angiotensin-
113.301811. (1-7) prevents cardiac fibrosis in DOCA-salt model of hyperten-
47. Xiao L, Gao L, Lazartigues E, Zucker IH. Brain-selective overex- sion. Am J Physiol Heart Circ Physiol. 2006;290(6):H2417–23.
pression of angiotensin-converting enzyme 2 attenuates sympa- https://doi.org/10.1152/ajpheart.01170.2005.
thetic nerve activity and enhances baroreflex function in chronic 59. Xue B, Zhang Z, Johnson RF, Guo F, Hay M, Johnson AK.
heart failure. Hypertension. 2011;58(6):1057–65. https://doi.org/ Central endogenous angiotensin-(1-7) protects against
10.1161/HYPERTENSIONAHA.111.176636. aldosterone/NaCl-induced hypertension in female rats. Am J
48.• Guimaraes PS, Oliveira MF, Braga JF, Nadu AP, Schreihofer A, Physiol Heart Circ Physiol. 2013;305(5):H699–705. https://doi.
Santos RAS, et al. Increasing angiotensin-(1–7) levels in the brain org/10.1152/ajpheart.00193.2013.
attenuates metabolic syndrome-related risks in fructose-fed rats. 60. Meng W, Zhao W, Zhao T, Liu C, Chen Y, Liu H, et al. Autocrine
Hypertension. 2014;63(5):1078–1085. The authors demonstrat- and paracrine function of angiotensin 1-7 in tissue repair during
ed that the chronic increase of Ang-(1–7) levels in the brain hypertension. Am J Hypertens. 2014;27(6):775–82. https://doi.
promotes cardiovascular and metabolic effects that protect org/10.1093/ajh/hpt270.
animals with fructose-induced metabolic syndrome. https:// 61.• Guo L, Yin A, Zhang Q, Zhong T, O’Rourke ST, Sun C.
doi.org/10.1161/HYPERTENSIONAHA.113.01847. Angiotensin-(1–7) attenuates angiotensin II-induced cardiac hy-
49. Guimaraes PS, Santiago NM, Xavier CH, Velloso EPP, Fontes pertrophy via a Sirt3-dependent mechanism. Am J Physiol Heart
MAP, Santos RAS, et al. Chronic infusion of angiotensin-(1-7) Circ Physiol. 2017;312(5):H980–H991. In this paper it was
into the lateral ventricle of the brain attenuates hypertension in demonstrated that Ang-(1–7) significantly attenuates Ang II-
DOCA-salt rats. Am J Physiol Heart Circ Physiol. 2012;303(3): induced cardiac hypertrophy and perivascular fibrosis
H393–400. https://doi.org/10.1152/ajpheart.00075.2012. through of SOD2 expression via stimulation of Sirt3-
50.•• Santiago NM, Guimaraes PS, Sirvente RA, Oliveira LAM, dependent deacetylation of FoxO3a in cardiomyocytes.
Irigoyen MC, Santos RAS, et al. Lifetime overproduction of cir- https://doi.org/10.1152/ajpheart.00768.2016.
culating Angiotensin-(1–7) attenuates deoxycorticosterone 62. Lin L, Liu X, Xu J, Weng L, Ren J, Ge J, et al. Mas receptor
acetate-salt hypertension-induced cardiac dysfunction and remod- mediates cardioprotection of angiotensin-(1-7) against angiotensin
eling. Hypertension. 2010;55(4):889–896. In this work, it was II-induced cardiomyocyte autophagy and cardiac remodelling
demonstrated that animals with lifetime increase in circulat- through inhibition of oxidative stress. J Cell Mol Med.
ing levels of Ang-(1–7) are protected against the damage 2016;20(1):48–57. https://doi.org/10.1111/jcmm.12687.
caused by DOCA-salt hypertension model. https://doi.org/10. 63.•• Gomes ER, et al. Angiotensin-(1–7) prevents cardiomyocyte path-
1161/HYPERTENSIONAHA.110.149815. ological remodeling through a nitric oxide/guanosine 3′,5′-cyclic
51. Bertagnolli M, Casali KR, de Sousa FB, Rigatto K, Becker L, monophosphate-dependent pathway. Hypertension. 2010;55(1):
Santos SHS, et al. An orally active angiotensin-(1-7) inclusion 153–160. This paper demonstrated that the protective NO/
compound and exercise training produce similar cardiovascular cGMP signaling pathway is activated by Ang-(1–7) on Ang
effects in spontaneously hypertensive rats. Peptides. 2014;51: II-induced cardiomyocyte remodeling. https://doi.org/10.1161/
65–73. https://doi.org/10.1016/j.peptides.2013.11.006. HYPERTENSIONAHA.109.143255.
52. Lu W, Kang J, Hu K, Tang S, Zhou X, Yu S, et al. Angiotensin-(1- 64. Bennion DM, Haltigan E, Regenhardt RW, Steckelings UM,
7) relieved renal injury induced by chronic intermittent hypoxia in Sumners C. Neuroprotective mechanisms of the ACE2-angioten-
rats by reducing inflammation, oxidative stress and fibrosis. Braz J sin-(1-7)-Mas axis in stroke. Curr Hypertens Rep. 2015;17(2):3.
Med Biol Res. 2017;50(1):e5594. https://doi.org/10.1590/1414- https://doi.org/10.1007/s11906-014-0512-2.
431X20165594. 65. Jiang T, Gao L, Lu J, Zhang YD. ACE2-Ang-(1-7)-Mas axis in
53. Li P, Zhang F, Sun HJ, Zhang F, Han Y. Angiotensin-(1-7) en- brain: a potential target for prevention and treatment of ischemic
hances the effects of angiotensin II on the cardiac sympathetic stroke. Curr Neuropharmacol. 2013;11(2):209–17. https://doi.org/
afferent reflex and sympathetic activity in rostral ventrolateral me- 10.2174/1570159X11311020007.
dulla in renovascular hypertensive rats. J Am Soc Hypertens. 66.• Regenhardt RW, Mecca AP, Desland F, Ritucci-Chinni PF, Ludin
2015;9(11):865–77. https://doi.org/10.1016/j.jash.2015.08.005. JA, Greenstein D, et al. Centrally administered angiotensin-(1–7)
54. Du D, Chen J, Liu M, Zhu M, Jing H, Fang J, et al. The effects of increases the survival of stroke-prone spontaneously hypertensive
angiotensin II and angiotensin-(1-7) in the rostral ventrolateral rats. Exp Physiol. 2014;99(2):442–453. These authors showed
medulla of rats on stress-induced hypertension. PLoS One. the beneficial effects of central administration of Ang-(1–7) on
2013;8(8):e70976. https://doi.org/10.1371/journal.pone.0070976. s t rok e- pro ne s po nt a neo us ly h yp erte nsi ve rat s , a
55. Oscar CG, Müller-Ribeiro FCF, de Castro LG, Martins Lima A, haemorrhagic stroke model, demonstrating its therapeutic po-
Campagnole-Santos MJ, Santos RAS, et al. Angiotensin-(1-7) in tential in this disease. https://doi.org/10.1113/expphysiol.2013.
the basolateral amygdala attenuates the cardiovascular response 075242.
evoked by acute emotional stress. Brain Res. 2015;1594:183–9. 67. Cunha TM, et al. The nonpeptide ANG-(1-7) mimic AVE 0991
https://doi.org/10.1016/j.brainres.2014.11.006. attenuates cardiac remodeling and improves baroreflex sensitivity
56. Olivon VC, Aires RD, Santiago LB, Ramalho LZN, Cortes SF, in renovascular hypertensive rats. Life Sci. 2013;92(4–5):266–75.
Lemos VS. Mas receptor overexpression increased Ang-(1-7) https://doi.org/10.1016/j.lfs.2012.12.008.
17 Page 14 of 15 Curr Hypertens Rep (2018) 20: 17

68. Souza AP, et al. Angiotensin II type 1 receptor blockade restores 83. Paul M, Wagner J, Dzau VJ. Gene expression of the renin-
angiotensin-(1-7)-induced coronary vasodilation in hypertrophic angiotensin system in human tissues. Quantitative analysis by
rat hearts. Clin Sci (Lond). 2013;125(9):449–59. https://doi.org/ the polymerase chain reaction. J Clin Invest. 1993;91(5):2058–
10.1042/CS20120519. 64. https://doi.org/10.1172/JCI116428.
69. Raffai G, Durand MJ, Lombard JH. Acute and chronic angioten- 84. Agoudemos MM, Greene AS. Localization of the renin-
sin-(1-7) restores vasodilation and reduces oxidative stress in mes- angiotensin system components to the skeletal muscle microcir-
enteric arteries of salt-fed rats. Am J Physiol Heart Circ Physiol. culation. Microcirculation. 2005;12(8):627–36. https://doi.org/10.
2011;301(4):H1341–52. https://doi.org/10.1152/ajpheart.00202. 1080/10739680500301664.
2011. 85. Zulli A, Burrell LM, Widdop RE, Black MJ, Buxton BF, Hare DL.
70. Savergnini SQ, Beiman M, Lautner RQ, de Paula-Carvalho V, Immunolocalization of ACE2 and AT2 receptors in rabbit athero-
Allahdadi K, Pessoa DC, et al. Vascular relaxation, antihyperten- sclerotic plaques. J Histochem Cytochem. 2006;54(2):147–50.
sive effect, and cardioprotection of a novel peptide agonist of the https://doi.org/10.1369/jhc.5C6782.2005.
MAS receptor. Hypertension. 2010;56(1):112–20. https://doi.org/ 86.•• Tesanovic S, Vinh A, Gaspari TA, Casley D, Widdop RE.
10.1161/HYPERTENSIONAHA.110.152942. Vasoprotective and atheroprotective effects of angiotensin (1–7)
71. Savergnini SQ, Ianzer D, Carvalho MBL, Ferreira AJ, Silva GAB, in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc
Marques FD, et al. The novel Mas agonist, CGEN-856S, attenu- Biol. 2010;30(8):1606–1613. First published paper on the role
ates isoproterenol-induced cardiac remodeling and myocardial in- of Ang-(1–7) in experimental atherosclerosis using ApoE
farction injury in rats. PLoS One. 2013;8(3):e57757. https://doi. knockout mice model, showing that the renin-angiotensin sys-
org/10.1371/journal.pone.0057757. tem is able to delay the progression of atherosclerosis. https://
72. Tetzner A, Gebolys K, Meinert C, Klein S, Uhlich A, Trebicka J, doi.org/10.1161/ATVBAHA.110.204453.
et al. G-protein-coupled receptor MrgD is a receptor for angioten- 87. Dong B, Zhang C, Feng JB, Zhao YX, Li SY, Yang YP, et al.
sin-(1-7) involving adenylyl cyclase, cAMP, and phosphokinase Overexpression of ACE2 enhances plaque stability in a rabbit
A. Hypertension. 2016;68(1):185–94. https://doi.org/10.1161/ model of atherosclerosis. Arterioscler Thromb Vasc Biol.
HYPERTENSIONAHA.116.07572. 2008;28(7):1270–6. https://doi.org/10.1161/ATVBAHA.108.
73. Soares ER, Barbosa CM, Campagnole-Santos MJ, Santos RAS, 164715.
Alzamora AC. Hypotensive effect induced by microinjection of
88. Lovren F, Pan Y, Quan A, Teoh H, Wang G, Shukla PC, et al.
Alamandine, a derivative of angiotensin-(1-7), into caudal ventro-
Angiotensin converting enzyme-2 confers endothelial protection
lateral medulla of 2K1C hypertensive rats. Peptides. 2017;96:67–
and attenuates atherosclerosis. Am J Physiol Heart Circ Physiol.
75. https://doi.org/10.1016/j.peptides.2017.09.005.
2008;295(4):H1377–84. https://doi.org/10.1152/ajpheart.00331.
74. Soltani Hekmat A, Javanmardi K, Kouhpayeh A, Baharamali E,
2008.
Farjam M. Differences in cardiovascular responses to alamandine
89. Zhang YH, Zhang Yh, Dong XF, Hao QQ, Zhou XM, Yu QT,
in two-kidney, one clip hypertensive and normotensive rats. Circ J.
et al. ACE2 and Ang-(1-7) protect endothelial cell function and
2017;81(3):405–12. https://doi.org/10.1253/circj.CJ-16-0958.
prevent early atherosclerosis by inhibiting inflammatory response.
75.• Anitschkow N, Chalatow S. Ueber experimentelle Cholester-
Inflamm Res. 2015;64(3–4):253–60. https://doi.org/10.1007/
insteatose und ihre Bedeutung fuer die Entstehung einiger
s00011-015-0805-1.
pathologischer Prozesse. Zentrbl Allg Pathol Pathol Anat.
1913;24:1–9. First published paper on animal model for ath- 90. Sluimer JC, Gasc JM, Hamming I, van Goor H, Michaud A, van
erosclerosis, from which further investigation emerged and, den Akker LH, et al. Angiotensin-converting enzyme 2 (ACE2)
thus, helped to conceive new approaches in animal models, expression and activity in human carotid atherosclerotic lesions. J
to improve experimental designs and to elucidate its Pathol. 2008;215(3):273–9. https://doi.org/10.1002/path.2357.
pathophysiology. 91. Zhang C, Zhao YX, Zhang YH, Zhu L, Deng BP, Zhou ZL, et al.
76. Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, García- Angiotensin-converting enzyme 2 attenuates atherosclerotic le-
Cardeña G, et al. Recommendation on design, execution, and sions by targeting vascular cells. Proc Natl Acad Sci U S A.
reporting of animal atherosclerosis studies: a scientific statement 2010;107(36):15886–91. https://doi.org/10.1073/pnas.
from the American Heart Association. Circ Res. 2017;121(6): 1001253107.
e53–79. https://doi.org/10.1161/RES.0000000000000169. 92.•• Fraga-Silva RA, Savergnini SQ, Montecucco F, Nencioni A,
77. Emini Veseli B, Perrotta P, de Meyer GRA, Roth L, van der Caffa I, Soncini D, et al. Treatment with angiotensin-(1–7) re-
Donckt C, Martinet W, et al. Animal models of atherosclerosis. duces inflammation in carotid atherosclerotic plaques. Thromb
Eur J Pharmacol. 2017;816:3–13. https://doi.org/10.1016/j.ejphar. Haemost. 2014;111(4):736–747. This paper delineates the ef-
2017.05.010. fects of Ang-(1–7)/HPβCD orally treated ApoE knockout
78. Lee YT, Lin HY, Chan YWF, Li KHC, To OTL, Yan BP, et al. mice on different shear stress prone regions, adding new data
Mouse models of atherosclerosis: a historical perspective and re- about Ang-(1–7) mechanisms in atherosclerotic plaques.
cent advances. Lipids Health Dis. 2017;16(1):12. https://doi.org/ https://doi.org/10.1160/TH13-06-0448.
10.1186/s12944-016-0402-5. 93. Silva AR, Aguilar EC, Alvarez-Leite JI, da Silva RF, Arantes
79. Bader M, Ganten D. Update on tissue renin-angiotensin systems. J RME, Bader M, et al. Mas receptor deficiency is associated with
Mol Med (Berl). 2008;86(6):615–21. https://doi.org/10.1007/ worsening of lipid profile and severe hepatic steatosis in ApoE-
s00109-008-0336-0. knockout mice. Am J Phys Regul Integr Comp Phys.
80. Sata M, Fukuda D. Crucial role of renin-angiotensin system in the 2013;305(11):R1323–30. https://doi.org/10.1152/ajpregu.00249.
pathogenesis of atherosclerosis. J Med Investig. 2010;57(1–2):12– 2013.
25. https://doi.org/10.2152/jmi.57.12. 94.• Hammer A, Yang G, Friedrich J, Kovacs A, Lee DH, Grave K,
81. Oliver JA, Sciacca RR. Local generation of angiotensin II as a et al. Role of the receptor Mas in macrophage-mediated inflam-
mechanism of regulation of peripheral vascular tone in the rat. J mation in vivo. Proc Natl Acad Sci U S A. 2016;113(49):14109–
Clin Invest. 1984;74(4):1247–51. https://doi.org/10.1172/ 14114. A new approach, by combining knockout models (Mas
JCI111534. receptor and ApoE), allowed further investigation on the role
82. Swales JD, Samani NJ. Vascular RAA system. J Hum Hypertens. of Mas receptor in sponteneously atherosclerotic mice, which
1993;7(Suppl 2):S3–6. lack ApoE expression, as well as the dynamics of these two
Curr Hypertens Rep (2018) 20: 17 Page 15 of 15 17

important components in ahterosclerosis. https://doi.org/10. 102.• Skiba DS, et al. Anti-atherosclerotic effect of the angiotensin 1–7
1073/pnas.1612668113. mimetic AVE0991 is mediated by inhibition of perivascular and
95. Yang JM, Dong M, Meng X, Zhao YX, Yang XY, Liu XL, et al. plaque inflammation in early atherosclerosis. Br J Pharmacol.
Angiotensin-(1-7) dose-dependently inhibits atherosclerotic lesion 2016; Newest paper about an Ang-(1–7) analogue, AVE0991,
formation and enhances plaque stability by targeting vascular and its role in atherosclerosis. This paper comprises previous-
cells. Arterioscler Thromb Vasc Biol. 2013;33(8):1978–85. ly published works on that issue and provides further infor-
https://doi.org/10.1161/ATVBAHA.113.301320. mation on mechanisms underlying the role of AVE0991.
96. Zhang F, Li S, Song J, Liu J, Cui Y, Chen H. Angiotensin-(1-7) 103. Habiyakare B, Alsaadon H, Mathai ML, Hayes A, Zulli A.
regulates angiotensin II-induced matrix metalloproteinase-8 in Reduction of angiotensin A and alamandine vasoactivity in the
vascular smooth muscle cells. Atherosclerosis. 2017;261:90–8. rabbit model of atherogenesis: differential effects of alamandine
https://doi.org/10.1016/j.atherosclerosis.2017.02.012. and Ang(1-7). Int J Exp Pathol. 2014;95(4):290–5. https://doi.org/
97. Bader M, Alenina N, Andrade-Navarro MA, Santos RA. MAS 10.1111/iep.12087.
and its related G protein-coupled receptors,Mrgprs. Pharmacol 104.•• Da Silva AR, Lenglet S, Carbone F, Burger F, Roth A, Liberale L,
Rev. 2014;66(4):1080–105. https://doi.org/10.1124/pr.113. et al. Alamandine abrogates neutrophil degranulation in athero-
008136. sclerotic mice. Eur J Clin Investig. 2017;47(2):117–128. First
98. Toton-Zuranska J, Gajda M, Pyka-Fosciak G, Kus K, Pawlowska published paper on the role of alamandine in experimental
M, Niepsuj A, et al. AVE 0991-angiotensin-(1-7) receptor agonist, atherosclerosis, extending the anti-atherosclerotic effects of
inhibits atherogenesis in apoE-knockout mice. J Physiol the protective arm of the renin-angiotensin system to its
Pharmacol. 2010;61(2):181–3. newest characterized component. https://doi.org/10.1111/eci.
99. Jawien J, Toton-Zuranska J, Kus K, Pawlowska M, Olszanecki R, 12708.
Korbut R. The effect of AVE 0991, nebivolol and doxycycline on 105.• Uchiyama T, Okajima F, Mogi C, Tobo A, Tomono S, Sato K.
inflammatory mediators in an apoE-knockout mouse model of Alamandine reduces leptin expression through the c-Src/p38
atherosclerosis. Med Sci Monit. 2012;18(10):Br389–93. MAP kinase pathway in adipose tissue. PLoS One. 2017;12(6):
100. Olszanecki R, Suski M, Gebska A, Toton-Zuranska J, Kus K, e0178769. This paper brings interesting data on the effects of
Madej J, et al. The influence of angiotensin-(1-7) peptidomimetic alamandine on plasminogen activator inhibitor-1 (PAI-1),
(AVE 0991) and nebivolol on angiotensin I metabolism in aorta of which is a pro-atherogenic protein. However, it leaves unan-
apoE-knockout mice. J Physiol Pharmacol. 2013;64(3):317–20. swered questions whether alamandine is a pro- or an anti-
101. Jawien J, Toton-Zuranska J, Gajda M, Niepsuj A, Gebska A, Kus atherogenic species. https://doi.org/10.1371/journal.pone.
K, et al. Angiotensin-(1-7) receptor Mas agonist ameliorates prog- 0178769.
ress of atherosclerosis in apoE-knockout mice. J Physiol
Pharmacol. 2012;63(1):77–85.
Current Hypertension Reports is a copyright of Springer, 2018. All Rights Reserved.

You might also like