ArticlePDF AvailableLiterature Review

Abstract and Figures

Hexarelin, a synthetic growth hormone-releasing peptide, can bind to and activate the growth hormone secretagogue receptor (GHSR) in the brain similar to its natural analog ghrelin. However, the peripheral distribution of GHSR in the heart and blood vessels suggests that hexarelin might have direct cardiovascular actions beyond growth hormone release and neuroendocrine effects. Furthermore, the non-GHSR CD36 had been demonstrated to be a specific cardiac receptor for hexarelin and to mediate its cardioprotective effects. When compared with ghrelin, hexarelin is chemically more stable and functionally more potent. Therefore, it may be a promising therapeutic agent for some cardiovascular conditions. In this concise review, we discuss the current evidence for the cardiovascular action of hexarelin.
Content may be subject to copyright.
Journal of Geriatric Cardiology (2014) 11: 253258
©2014 JGC All rights reserved; www.jgc301.com
http://www.jgc301.com; jgc@jgc301.com | Journal of Geriatric Cardiology
Review Open Access
The cardiovascular action of hexarelin
Yuanjie MAO1,2, Takeshi Tokudome1, Ichiro Kishimoto1,3
1Department of Biochemistry, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
2Department of Medicine, Prince George’s Hospital Center, Cheverly, Maryland 20785, USA
3Department of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
Abstract
Hexarelin, a synthetic growth hormone-releasing peptide, can bind to and activate the growth hormone secretagogue receptor (GHSR)
in the brain similar to its natural analog ghrelin. However, the peripheral distribution of GHSR in the heart and blood vessels suggests that
hexarelin might have direct cardiovascular actions beyond growth hormone release and neuroendocrine effects. Furthermore, the non-GHSR
CD36 had been demonstrated to be a specific cardiac receptor for hexarelin and to mediate its cardioprotective effects. When compared with
ghrelin, hexarelin is chemically more stable and functionally more potent. Therefore, it may be a promising therapeutic agent for some car-
diovascular conditions. In this concise review, we discuss the current evidence for the cardiovascular action of hexarelin.
J Geriatr Cardiol 2014; 11: 253258. doi:10.11909/j.issn.1671-5411.2014.03.007
Keywords: Hexarelin; Cardiovascular disease; Growth hormone secretagogue receptor; CD36
1 Introduction
Growth hormone secretagogues (GHS) are a class of
small synthetic peptides that stimulate growth hormone (GH)
release through binding to the growth hormone secre-
tagogue receptor (GHSR) 1a. Moreover, GHSR 1a is a
G-protein-coupled receptor originally identified in the hy-
pothalamus and pituitary,[1] and later recognized as the re-
ceptor for the endogenous hormone ghrelin.[2] The periph-
eral distribution of GHSR 1a in the heart, adrenals, fat,
prostate, bone, and digestive tract has supported physio-
logical roles of GHSs and ghrelin independent of GH re-
lease and neuroendocrine stimulation. For example, GH-in-
dependent effects on orexigenic properties, fat metabolism,
immune, gastrointestinal, and cardiovascular activities have
been reported for GHSs and ghrelin.[3–6 ]
Previous studies have revealed that ghrelin administra-
tion can improve cardiac function in rats and patients with
chronic heart failure, as indicated by increased left ventricle
ejection fraction (LVEF), cardiac output, and exercise ca-
pacity.[7–9 ] In rodents with acute myocardial infarction (MI),
ghrelin administration prevented malignant arrhythmias and
reduced mortality in the acute phase, while improving left
Correspondence to: Ichiro Kishimoto, MD, Department of Biochemistry,
National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fuji-
shiro-dai, Suita, Osaka 565-8565, Japan. E-mail: kishimot@hsp.ncvc.go.jp
Telephone: +81-668-335015 Fax: +81-668-355402
Received: April 2, 2014 Revised: May 25, 2014
Accepted: July 10, 2014 Published online: September 10, 2014
ventricle (LV) dysfunction and attenuating cardiac remod-
eling in the subacute phase.[10–13] However, ghrelin is an
unstable natural peptide that is transformed and degraded,
which limits its clinical use. The GHS hexarelin is a chemi-
cally stable and potent synthetic hexapeptide that can be
administered orally, making it a potential alternative to
ghrelin.[14] It is comparable to ghrelin with respect to the
half-maximal effective concentration for their common re-
ceptor, GHSR 1a; although the cardiac action of hexarelin
was reported to be mediated in part by GHSR 1a and largely
by activation of the CD36 receptor, in isolated working
hearts.[15,16] In this concise review, we discuss the current
evidence for the cardiovascular action of hexarelin.
2 Cardiovascular action
2.1 Inotropic effect
Acute intravenous administration of hexarelin had a
short-lasting, positive inotropic effect. Cardiac performance
was studied by radionuclide angiocardiography in seven
male volunteers. Hexarelin administration increased LVEF
(70.7 ± 3.0% vs. 64.0 ± 1.5%, P < 0.03) without affecting
mean blood pressure and heart rate. LVEF was significantly
increased after 15 min and peaked at 30 min, and the effect
lasted for up to 60 min after administration.[17] In 24 male
patients with coronary artery disease undergoing by-pass
surgery under general anesthesia, LVEF, cardiac output,
and cardiac index were evaluated by transoesophageal
echocardiography while wedge pressure, central venous
254 MAO YJ, et al. Cardiovascular action of hexarelin
Journal of Geriatric Cardiology | jgc@jgc301.com; http://www.jgc301.com
pressure, mean arterial pressure, and systemic vascular re-
sistance index were determined by systemic and pulmonary
arterial catheterization. Acute intravenous administration
of hexarelin (2 µg/kg) induced a rapid increase in LVEF,
cardiac output, and cardiac index, while reducing wedge
pressure. It also increased mean arterial pressure and tran-
siently decreased central venous pressure, but did not
change the systemic vascular resistance index and heart
rate.[18] Furthermore, hexarelin induced time- and concen-
tration-dependent inotropic effects in rat papillary muscle,[19]
and increased the amplitude of intracellular Ca2+ transients
and L-type Ca2+ current to produce positive inotropic effects
in freshly isolated adult Wistar rat ventricular myocytes
through protein kinase C signaling cascade.[20].
2.2 Inhibition of apoptosis
Treatment of neonatal rat cardiomyocytes with hexarelin
significantly decreased angiotensin II-induced apoptosis and
DNA fragmentation, and increased myocyte viability.[21]
Hexarelin treatment also inhibited doxorubicin-induced
apoptosis and promoted survival of H9c2 cardiomyocytes
and endothelial cells.[22] The anti-apoptosis activity of hex-
arelin in cardiomyocytes and endothelial cells may partially
explain its cardioprotective effects. Chronic administration
of hexarelin alleviates LV dysfunction, pathological remod-
eling, and cardiac cachexia in rats with congestive heart
failure by suppressing stress-induced neurohormonal activa-
tion and cardiomyocyte apoptosis.[23]
2.3 Ischemia-reperfusion injury
In hearts subjected to 30 min of ischemia followed by
120 min of reperfusion, hexarelin (1 µmol/L) significantly
reduced infarct size, as determined by using triphenyltetra-
zolium chloride staining, and the protection provided by
hexarelin was partly abolished by the protein kinase C in-
hibitor chelerythrine.[24] Hexarelin treatment not only pre-
served the electrophysiological properties of cardiomyo-
cytes after ischemia-reperfusion injury but also inhibited
cardiomyocyte apoptosis and promoted cell survival by
modification of mitogen-activated protein kinase path-
ways,[25] and produced a positive inotropic effect on
ischemic cardiomyocytes.[26] Hexarelin administration for
30 days counteracted the ischemic heart damage in Zucker
rats subjected to low flow ischemia and reperfusion. The
recovery of LV pressure developed at reperfusion was sig-
nificantly greater in hexarelin-treated rats than in controls
and the increase in coronary resistance was minimal.[27] The
chronic administration of hexarelin to GH-deficient rats had
a pronounced protective effect against ischemic and
post-ischemic ventricular dysfunction, and prevented hy-
per-responsiveness of the coronary vascular bed to angio-
tensin II in perfused hearts.[28]
2.4 Myocardial infarction
Four weeks after ligation of the left coronary artery, male
rats were treated with hexarelin (100 µg/kg per day) or
normal saline subcutaneously for two weeks. Transthoracic
echocardiography was performed before and after the
treatment period. Compared with normal saline, hexarelin
treatment increased stroke volume, stroke volume in-
dex, cardiac output, and cardiac index, and decreased total
peripheral resistance.[29]
2.5 Cardiac fibrosis
Hexarelin treatment of spontaneously hypertensive rats
for five weeks significantly reduced cardiac fibrosis by de-
creasing interstitial and perivascular myocardial collagen
deposition and myocardial hydroxyproline content, and re-
ducing collagen I and III mRNA and protein expression. In
addition, hexarelin treatment increased matrix metallopro-
teinase-2 and -9 activities and decreased myocardial mRNA
expression of the tissue inhibitor of metalloproteinase-1.
Furthermore, hexarelin treatment significantly attenuated
LV hypertrophy, LV diastolic dysfunction, and high blood
pressure.[30] Treatment of cultured cardiac fibroblasts with
hexarelin (0.1 µmol/L) inhibited angiotensin II-induced
proliferation and collagen synthesis, and transforming
growth factor (TGF)-β-induced DNA synthesis, and re-
duced the angiotensin II-mediated upregulation of TGF-β
expression and release.[31]
2.6 Atherosclerosis
Anti-atherosclerotic activity of hexarelin was observed in
adult Sprague-Dawley rats. Treatment with hexarelin sup-
pressed the formation of atherosclerotic plaques and neoin-
tima, partially reversed serum high-density lipoprotein cho-
lesterol/low-density lipoprotein cholesterol ratio, and in-
creased serum nitric oxide levels and aortic mRNA expres-
sion of endothelial nitric oxide synthase, GHSRs, and CD36
in atherosclerotic rats. Hexarelin treatment also decreased
tritiated thymidine incorporation in cultured vascular smooth
muscle cells, calcium sedimentation in the aortic wall, and
foam cell formation induced by oxidized low-density lipo-
protein.[32] Furthermore, chronic treatment with hexarelin
unaltered the high triglyceride levels and significantly de-
creased plasma cholesterol concentrations in obese rats.[27]
3 Cardiac receptor
The cardiovascular action of hexarelin has been regarded
MAO YJ, et al. The cardiovascular action of hexarelin 255
http://www.jgc301.com; jgc@mail.sciencep.com | Journal of Geriatric Cardiology
as GH-independent and occurs through activation of cardiac
receptors. Previous studies showed that the cardiovascular
effects of hexarelin are not shared by recombinant human
GH or by GH-releasing hormone, indicating that they are
not mediated by an increase in circulating GH levels.[17,18,33]
Moreover, hexarelin significantly increased LVEF in nor-
mal and in GH-deficient patients[34–36] and prevented cardiac
damage after ischemia-reperfusion in hypophysectomized
rats,[37] indicating that its cardioprotective activity is not due
to stimulation of the GH axis.[27]
Hexarelin can bind to specific cardiac sites. Specific
125I-Tyr-Ala-hexarelin binding was observed in the human
cardiovascular system, and the highest 125I-Tyr-Ala-hex-
arelin levels were detected in the ventricles, followed by
atria, aorta, coronaries, carotid, endocardium, and vena ca-
va.[38] Specific hexarelin binding has also been shown in
H9c2 myocytes.[39] Currently, two cardiac receptor subtypes
have been proposed for hexarelin.
3.1 Cardiac GHSR 1a receptor
GHSR mRNA expression in cardiomyocytes was upre-
gulated after treatment with hexarelin,[21] and GHSR 1a
protein was expressed primarily in the heart as compared to
all other organs.[40] Fluorescein-conjugated ghrelin (1–18)
bound specifically to heart tissue in situ and was displaced
by both excess ghrelin and hexarelin.[40] Further, hexarelin
significantly prolonged action potential duration, produced
positive inotropic effects, and preserved electrophysiologi-
cal properties after ischemia-reperfusion injury in isolated
myocytes. These effects were abolished in the presence of
the GHSR antagonist d-Lys-3-GH-releasing peptide-6 or the
GHSR 1a-specific antagonist BIM28163.[20,25,26,41] The ef-
fects of hexarelin on cardiac function, cardiac fibrosis, and
blood pressure were also mediated by GHSRs, since GHSR
expression was upregulated by hexarelin treatment and a
selective GHSR antagonist inhibited hexarelin activity.[30]
3.2 Cardiac CD36 receptor
The presence of specific GHS binding sites was demon-
strated in three different human breast carcinoma cell lines
(MCF7, T47D, and MDA-MB-231), which lacked detect-
able GHSR 1a mRNA expression. However, hexarelin
treatment significantly inhibited proliferation of these cell
lines at concentrations close to the binding affinity.[42] A
photoactivatable derivative of hexarelin was developed to
label and characterize binding sites in anterior pituitary
membranes. The differential binding affinity for car-
diac tissue raised the possibility of the existence of distinct
receptor subtypes in the pituitary and the cardiovascular
system.[43] GHSRs were detected mainly in the myocardium
by using a radioreceptor assay with 125I-Tyr-Ala-hexarelin,
but they were also present in the adrenals, gonads, arteries,
lungs, liver, skeletal muscle, kidneys, pituitary, thyroid,
adipose tissue, veins, uterus, skin, and lymph nodes. Hex-
arelin and human ghrelin completely displaced the radioli-
gand from binding sites in endocrine tissues, but ghrelin was
less potent than hexarelin. In non-endocrine tissues, such as
heart, ghrelin did not displace 125I-Tyr-Ala-hexarelin,
whereas hexarelin had the same displacement activity as in
endocrine tissues. This suggested that there is a hex-
arelin-specific receptor subtype in the heart and in other
non-endocrine tissues.[44] Finally, the specific cardiac re-
ceptor for hexarelin was identified. The N-terminal se-
quence of the deglycosylated protein was identical to rat
CD36, a multifunctional glycoprotein, which is expressed in
cardiomyocytes and microvascular endothelial cells. Hex-
arelin-mediated activation of CD36 in perfused hearts in-
creased coronary perfusion pressure in a dose-dependent
manner. This effect was not observed in hearts from
CD36-null mice and from spontaneously hypertensive rats
genetically deficient in CD36.[45, 46]
4 Hexarelin vs. ghrelin
Hexarelin has more potent beneficial effects on the car-
diovascular system compared with its natural analog ghrelin.
In one study, either ghrelin (320 μg/kg per day) or equimo-
lar hexarelin (80 μg/kg per day) was administered to hy-
pophysectomized rats for seven days and their hearts were
then subjected to ischemia and reperfusion in vitro. Hex-
arelin was more potent than ghrelin in preventing increases
in LV end-diastolic pressure, coronary perfusion pressure,
and creatine kinase release in the heart perfusate.[15] In an-
other study, chronic hexarelin administration improved
heart function in ghrelin-null mice to a greater extent than
equimolar ghrelin administration after experimental MI.[47]
Given the fact that the half-maximal effective concentration
of hexarelin for GHSR 1a (1.7 nmol/L) is comparable to
that of ghrelin (1.0 nmol/L),[16] the higher potency of hex-
arelin was considered to be mediated largely by interactions
with CD36 in the heart, and in part by GHSRs.[15,47]
However, other studies reported that when GHSR 1a ac-
tivation was identical hexarelin and ghrelin had similar car-
diac effects, although the dosage of ghrelin was 10 times
higher than that of hexarelin in molar terms. Ghrelin (10
nmol/L) or hexarelin (1 nmol/L) addition to the perfusion
system after ischemia had a positive inotropic effect on
ischemic cardiomyocytes through activation of the GHSR
1a receptor, thereby protecting them from ischemia-re-
perfusion injury.[20,26] Another study suggested that ghrelin-
256 MAO YJ, et al. Cardiovascular action of hexarelin
Journal of Geriatric Cardiology | jgc@jgc301.com; http://www.jgc301.com
and hexarelin-mediated activation of GHSR 1a had a similar
protective effect on cardiomyocytes after ischemia-reper-
fusion injury by inhibiting cardiomyocyte apoptosis and
promoting cell survival.[25] The common features of the two
peptides were compared in Table 1.
5 Conclusions
Hexarelin has cardioprotective activity in common car-
diovascular conditions such as cardiac fibrosis, ischemic
heart disease, cardiac dysfunction, and atherosclerosis. The
important in vivo studies of hexarelin in cardiovascular con-
ditions are summarized in Table 2. These beneficial effects
seem to be mediated through the direct binding and activa-
tion of its cardiac receptors CD36 and GHSR 1a. Since
hexarelin is a chemically stable synthetic GHS with more
potent cardiac effects than its natural analog ghrelin, it can
be a potential alternative to ghrelin as a promising
Table 1. Comparison of hexarelin and ghrelin.
Hexarelin Ghrelin
Source Synthetic Natural
Chemical structure 6 Amino acids 28 Amino acids
Amino acid sequence His-D-2-methyl-Trp-Ala-Trp-
D-Phe-Lys-NH2
Gly-Ser-Ser(octanoyl)-Phe-Leu-Ser-Pro-Glu-His-Gln-Arg-Val-Gln-Gln-
Arg-Lys-Glu-Ser-Lys- Lys-Pro-Pro-Ala-Lys-Leu-Gln-Pro-Arg-OH
Half life 57–71 min[48] 11–17 min,[49] 27–31 min[50]
Receptor GHSR1a; CD36 GHSR1a
Receptor affinity for GHSR1a (EC50) 1.7 nmol/L[16] 1.0 nmol/L[16]
GHSR1a: growth hormone secretagogue receptor; EC50: half-maximal effective concentration.
Table 2. In vivo studies of the cardiovascular action of hexarelin.
First author, date Species Model Dose, duration, initiation of treatment Main outcomes
Mao, et al, 2013[47] Ghrelin-
null mice
Experimental myocardial infarction
by coronary artery ligation
300μg/kg per day for 14 days, from 30 min
after ligation (s.c.)
Improved heart failure
better than ghrelin
Xu, et al, 2012[30] Rats Spontaneous hypertension 100 μg/kg per day for 5 weeks, from an age o
f
16 weeks (s.c.) Reduced cardiac fibrosis
Pang, et al, 2010[32] Rats
High lipid diet and vitamin D3-
induced atherosclerosis
200 μg/kg per day for 30 days, in the last
month after high lipid diet (s.c.)
Alleviated the development
of atherosclerosis
Xu, et al, 2005 [23] Rats
Pressure-overload heart failure by
abdominal aortic banding
200 μg/kg per day for 3 weeks, from 9 weeks
after heart failure (s.c.)
Alleviated LV dysfunction, pathological
remodeling, and cardiac cachexia
Torsello, et al,
2003 [15] Rats Hypophysectomized 80 μg/kg per day for 7 days, before in vitro
ischemia and reperfusion procedure (s.c.)
Far more effective than ghrelin in the
control of heart function
Broglio, et al,
2002 [18] Humans Coronary artery disease during
by-pass surgery 2 μg/kg acute administration (i.v.) Increased LVEF, cardiac index
and cardiac output
Imazio, et al,
2002 [34] Humans Normal, dilated, and ischemic
cardiomyopathy 2 μg/kg acute administration (i.v.)
Increased LVEF in ischemic cardio-
myopathy patients and in normals but
not in dilated cardiomyopathy patients
Broglio, et al,
2001 [35] Humans
Normal adults, growth hormone-
deficient patients, and severe dilated
cardiomyopathy patients
2 μg/kg acute administration (i.v.) Produced a positive inotropic effect
De Gennaro-Colonna,
et al, 2000 [27]
Zucker
rats Obese 160 μg/kg per day for 30 days, at 30
weeks of age (s.c.)
Induced cardioprotective effect after is-
chemia and decreased plasma cholesterol
Tivesten, et al,
2000 [29] Rats Experimental myocardial infarction
by coronary artery ligation
10 μg/kg per day or 100μg/kg per day for 2
weeks, from 4 weeks after ligation (s.c.)
Improved cardiac function and
decreased peripheral resistance
Bisi, et al, 1999[36] Humans Growth hormone deficiency 2μg/kg acute administration (i.v.) Increased LVEF
Locatelli, et al,
1999[37] Rats Hypophysectomized 80 μg/kg per day for 7 days, be-
fore ischemia-reperfusion damage (s.c.)
Prevented cardiac damage after
ischemia-reperfusion
Bisi, et al, 1999[17] Humans Volunteers 2μg/kg acute administration (i.v.) Increased LVEF without significant chan-
ges in mean blood pressure and heart rate
De Gennaro Colonna,
et al, 1997[51] Rats Anti-GHRH serum-treated 160 μg/kg per day for 15 days, after administra-
tion of an anti-GHRH serum for 20 days (s.c.)Counteracted the ischemic damage
LV: left ventricle; LVEF: left ventricular ejection fraction; GHRH: growth hormone releasing hormone.
MAO YJ, et al. The cardiovascular action of hexarelin 257
http://www.jgc301.com; jgc@mail.sciencep.com | Journal of Geriatric Cardiology
therapeutic agent for the treatment of cardiovascular dis-
eases. However, as current evidence is mainly from experi-
mental animal models or in vitro cell lines, clinical trials
aimed to extend the application of hexarelin in human sub-
jects and observe its efficacy and potential side effects are
warranted.
References
1 Howard AD, Feighner SD, Cully DF, et al. A receptor in pi-
tuitary and hypothalamus that functions in growth hormone
release. Science 1996; 273: 974–977.
2 Kojima M, Hosoda H, Matsuo H, et al. Ghrelin: discovery of
the natural endogenous ligand for the growth hormone secre-
tagogue receptor. Trends Endocrinol Metab 2001; 12:
118–122.
3 Lazarczyk MA, Lazarczyk M, Grzela T. Ghrelin: a recently
discovered gut-brain peptide (review). Int J Mol Med 2003; 12:
279–287.
4 Marleau S, Mulumba M, Lamontagne D, et al. Cardiac and
peripheral actions of growth hormone and its releasing pep-
tides: relevance for the treatment of cardiomyopathies. Car-
diovasc Res 2006; 69: 26–35.
5 Leite-Moreira AF, Soares JB. Physiological, pathological and
potential therapeutic roles of ghrelin. Drug Discov Today
2007; 12: 276–288.
6 van der Lely AJ, Tschöp M, Heiman ML, et al. Biological,
physiological, pathophysiological, and pharmacological as-
pects of ghrelin. Endocr Rev 2004; 25: 426–457.
7 Nagaya N, Miyatake K, Uematsu M, et al. Hemodynamic,
renal, and hormonal effects of ghrelin infusion in patients with
chronic heart failure. J Clin Endocrinol Metab 2001; 86:
5854–5859.
8 Nagaya N, Moriya J, Yasumura Y, et al. Effects of ghrelin
administration on left ventricular function, exercise capacity,
and muscle wasting in patients with chronic heart failure.
Circulation 2004; 110: 3674–3679.
9 Nagaya N, Uematsu M, Kojima M, et al. Chronic administra-
tion of ghrelin improves left ventricular dysfunction and at-
tenuates development of cardiac cachexia in rats with heart
failure. Circulation 2001; 104: 1430-1435.
10 Soeki T, Kishimoto I, Schwenke DO, et al. Ghrelin suppresses
cardiac sympathetic activity and prevents early left ventricular
remodeling in rats with myocardial infarction. Am J Physiol
Heart Circ Physiol 2008; 294: H426-H432.
11 Mao Y, Tokudome T, Otani K, et al. Ghrelin prevents inci-
dence of malignant arrhythmia after acute myocardial infarc-
tion through vagal afferent nerves. Endocrinology 2012; 153:
3426–3434.
12 Schwenke DO, Tokudome T, Kishimoto I, et al. Early ghrelin
treatment after myocardial infarction prevents an increase in
cardiac sympathetic tone and reduces mortality. Endocrinol-
ogy 2008; 149: 5172–5176.
13 Mao Y, Tokudome T, Otani K, et al. Excessive sympathoac-
tivation and deteriorated heart function after myocardial in-
farction in male ghrelin knockout mice. Endocrinology 2013;
154: 1854–1863.
14 Deghenghi R, Cananzi MM, Torsello A, et al. GH-releasing
activity of hexarelin, a new growth hormone releasing peptide,
in infant and adult rats. Life Sci 1994; 54: 1321–1328.
15 Torsello A, Bresciani E, Rossoni G, et al. Ghrelin plays a
minor role in the physiological control of cardiac function in
the rat. Endocrinology 2003; 144:1787–1792.
16 Falls HD, Dayton BD, Fry DG, et al. Characterization of
ghrelin receptor activity in a rat pituitary cell line RC-4B/C. J
Mol Endocrinol 2006; 37: 51–62.
17 Bisi G, Podio V, Valetto MR, et al. Acute cardiovascular and
hormonal effects of GH and hexarelin, a synthetic GH-releas-
ing peptide, in humans. J Endocrinol Invest 1999; 22: 266–272.
18 Broglio F, Guarracino F, Benso A, et al. Effects of
acute hexarelin administration on cardiac performance in pa-
tients with coronary artery disease during by-pass surgery.
Eur J Pharmacol 2002; 448: 193–200.
19 Bedendi I, Gallo MP, Malan D, et al. Role of endothelial cells
in modulation of contractility induced by hexarelin in rat ven-
tricle. Life Sci 2001; 69: 2189–2201.
20 Sun Q, Ma Y, Zhang L, et al. Effects of GH secretagogues on
contractility and Ca2+ homeostasis of isolated adult rat ven-
tricular myocytes. Endocrinology 2010; 151: 4446–4454.
21 Pang JJ, Xu RK, Xu XB, et al. Hexarelin protects rat cardio-
myocytes from angiotensin II-induced apoptosis in vitro. Am J
Physiol Heart Circ Physiol 2004; 286: H1063- H1069.
22 Filigheddu N, Fubini A, Baldanzi G, et al. Hexarelin protects
H9c2 cardiomyocytes from doxorubicin-induced cell death.
Endocrine 2001; 14: 113–119.
23 Xu XB, Pang JJ, Cao JM, et al. GH-releasing peptides im-
prove cardiac dysfunction and cachexia and suppress
stress-related hormones and cardiomyocyte apoptosis in rats
with heart failure. Am J Physiol Heart Circ Physiol 2005; 289:
H1643- H1651.
24 Frascarelli S, Ghelardoni S, Ronca-Testoni S, et al. Effect of
ghrelin and synthetic growth hormone secretagogues in nor-
mal and ischemic rat heart. Basic Res Cardio 2003; 98:
401–405.
25 Ma Y, Zhang L, Launikonis BS, et al. Growth hormone se-
cretagogues preserve the electrophysiological properties of
mouse cardiomyocytes isolated from in vitro ische-
mia/reperfusion heart. Endocrinology 2012; 153: 5480–5490.
26 Ma Y, Zhang L, Edwards JN, et al. Growth hormone secre-
tagogues protect mouse cardiomyocytes from in vitro ische-
mia/reperfusion injury through regulation of intracellular cal-
cium. PLoS One 2012; 7: e35265.
27 De Gennaro-Colonna V, Rossoni G, Cocchi D, et al. Endo-
crine, metabolic and cardioprotective effects of hexarelin in
obese Zucker rats. J Endocrinol 2000; 166: 529–536.
28 Berti F, Müller E, De Gennaro Colonna V, et al. Hex-
arelin exhibits protective activity against cardiac ischaemia in
258 MAO YJ, et al. Cardiovascular action of hexarelin
Journal of Geriatric Cardiology | jgc@jgc301.com; http://www.jgc301.com
hearts from growth hormone-deficient rats. Growth Horm IGF
Res 1998; 8(Suppl B): S149–S152.
29 Tivesten A, Bollano E, Caidahl K, et al. The growth hormone
secretagogue hexarelin improves cardiac function in rats after
experimental myocardial infarction. Endocrinology 2000; 141:
60–66.
30 Xu X, Ding F, Pang J, et al. Chronic administration of hex-
arelin attenuates cardiac fibrosis in the spontaneously hyper-
tensive rat. Am J Physiol Heart Circ Physiol 2012; 303:
H703–H711.
31 Xu X, Pang J, Yin H, et al. Hexarelin suppresses cardiac fi-
broblast proliferation and collagen synthesis in rat. Am J Phy-
siol Heart Circ Physiol 2007; 293: H2952–H2958.
32 Pang J, Xu Q, Xu X, et al. Hexarelin suppresses high lipid diet
and vitamin D3-induced atherosclerosis in the rat. Pep-
tides 2010; 31: 630–638.
33 Rossoni G, De Gennaro Colonna V, Bernareggi M, et al. Pro-
tectant activity of hexarelin or growth hormone against posti-
schemic ventricular dysfunction in hearts from aged rats. J
Cardiovasc Pharmacol 1998; 32: 260–265.
34 Imazio M, Bobbio M, Broglio F, et al. GH-independent car-
diotropic activities of hexarelin in patients with severe left
ventricular dysfunction due to dilated and ischemic cardio-
myopathy. Eur J Heart Fail 2002; 4: 185–191.
35 Broglio F, Benso A, Valetto MR, et al. Growth hor-
mone-independent cardiotropic activities of growth hor-
mone-releasing peptides in normal subjects, in patients with
growth hormone deficiency, and in patients with idiopathic or
ischemic dilated cardiomyopathy. Endocrine 2001; 14:
105–108.
36 Bisi G, Podio V, Valetto MR, et al. Cardiac effects of hex-
arelin in hypopituitary adults. Eur J Pharmacol 1999; 381:
31–38.
37 Locatelli V, Rossoni G, Schweiger F, et al. Growth hor-
mone-independent cardioprotective effects of hexarelin in the
rat. Endocrinology 1999; 140: 4024–4031.
38 Muccioli G, Broglio F, Valetto MR, et al. Growth hor-
mone-releasing peptides and the cardiovascular system. Ann
Endocrinol (Paris) 2000; 61: 27–31.
39 Pettersson I, Muccioli G, Granata R, et al. Natural (ghrelin)
and synthetic (hexarelin) GH secretagogues stimulate H9c2
cardiomyocyte cell proliferation. J Endocrinol 2002; 175:
201–209.
40 McGirr R, McFarland MS, McTavish J, et al. Design and
characterization of a fluorescent ghrelin analog for imaging
the growth hormone secretagogue receptor 1a. Regul
Pept 2011; 172: 69–76.
41 Sun Q, Zang WJ, Chen C. Growth hormone secretagogues
reduce transient outward K+ current via phospholipase
C/protein kinase C signaling pathway in rat ventricular myo-
cytes. Endocrinology 2010; 151: 1228–1235.
42 Cassoni P, Papotti M, Ghè C, et al. Identification, characteri-
zation, and biological activity of specific receptors for natural
(ghrelin) and synthetic growth hormone secretagogues and
analogs in human breast carcinomas and cell lines. J Clin En-
docrinol Metab 2001; 86: 1738–1745.
43 Ong H, Bodart V, McNicoll N, et al. Binding sites for growth
hormone-releasing peptide. Growth Horm IGF Res 1998; 8
(Suppl B): S137–S140.
44 Papotti M, Ghè C, Cassoni P, et al. Growth hormone secre-
tagogue binding sites in peripheral human tissues. J Clin En-
docrinol Metab 2000; 85: 3803–3807.
45 Bodart V, Febbraio M, Demers A, et al. CD36 mediates the
cardiovascular action of growth hormone-releasing peptides in
the heart. Circ Res 2002; 90: 844–849.
46 Bodart V, Bouchard JF, McNicoll N, et al. Identification and
characterization of a new growth hormone-releasing peptide
receptor in the heart. Circ Res 1999; 85: 796–802.
47 Mao Y, Tokudome T, Kishimoto I, et al. Hexarelin treatment
in male ghrelin knockout mice after myocardial infarction.
Endocrinology 2013; 154: 3847–3854.
48 Roumi M, Marleau S, du Souich P, et al. Kinetics and disposi-
tion of hexarelin, a peptidic growth hormone secretagogue, in
rats. Drug Metab Dispos 2000; 28: 44–50.
49 Wu R, Zhou M, Cui X, et al. Ghrelin clearance is reduced at
the late stage of polymicrobial sepsis. Int J Mol Med 2003; 12:
777–781.
50 Akamizu T, Takaya K, Irako T, et al. Pharmacokinetics, safety,
and endocrine and appetite effects of ghrelin administration in
young healthy subjects. Eur J Endocrinol 2004; 150:
447–455.
51 De Gennaro Colonna V, Rossoni G, Bernareggi M, et al.
Hexarelin, a growth hormone-releasing peptide, discloses
protectant activity against cardiovascular damage in rats with
isolated growth hormone deficiency. Cardiologia 1997; 42:
1165–1172.
... For more than a decade, there has been a great interest in GHS alternatives to ghrelin as a therapy. Hexarelin is a stable synthetic GHS peptide analog of ghrelin that has been shown to have a wide range of cytoprotective and cardioprotective actions in vitro and in vivo, such as amelioration of LV dysfunction and remodeling in the setting of pressure overload and myocardial infarction (Berti et al., 1998;Filigheddu et al., 2001;Mao et al., 2013aMao et al., , 2014aMcdonald et al., 2020). Hexarelin was demonstrated to have greater potency than ghrelin treatment in improving LV function after myocardial infarction in ghrelin deficient mice, which was suggested to be due to the possible binding of hexarelin to cardiac GHS-R1a and CD36 (class B scavenger receptor involved in fatty acid uptake) receptors (Mao et al., 2013a). ...
... To date, there have been few investigations of the vascular effects of hexarelin, other than showing that it reduces apoptosis in ischemia (Mao et al., 2014a). Hexarelin-mediated dilation is shown in this study to be dependent on local GHS-R1a, not because of change in perfusion pressure, as mean arterial pressure did not change significantly after acute administration. ...
... Moreover, there is no specific therapy targeted to RV failure in PH. To demonstrate the utility of SR imaging approaches, which complement state-of-the-art cardiology techniques for such studies on the pathogenesis and prevention of PH, we briefly described herein how RV function is affected by chronic administration of a GHS peptide that is widely considered as a promising therapy to reduce neurohormonal activation and adverse remodeling in cardiovascular disease (Berti et al., 1998;Filigheddu et al., 2001;Mao et al., 2013aMao et al., , 2014aMcdonald et al., 2020). The novel and somewhat surprising findings in this study, even with limited sample sizes, are that chronic simultaneous administration of hexarelin with SuHx treatment to induce severe PH did not prevent the pathogenesis of RV hypertrophy or RV myocardial dysfunction in the young adult male rats. ...
Article
Full-text available
The majority of the conventional techniques that are utilized for investigating the pathogenesis of cardiovascular disease in preclinical animal models do not permit microlevel assessment of in situ cardiomyocyte and microvascular functions. Therefore, it has been difficult to establish whether cardiac dysfunction in complex multiorgan disease states, such as heart failure with preserved ejection fraction and pulmonary hypertension, have their origins in microvascular dysfunction or rather in the cardiomyocyte. Herein, we describe our approach of utilizing synchrotron radiation microangiography to, first, ascertain whether the growth hormone secretagogue (GHS) hexarelin is a vasodilator in the coronary circulation of normal and anesthetized Sprague-Dawley rats, and next investigate if hexarelin is able to prevent the pathogenesis of right ventricle (RV) dysfunction in pulmonary hypertension in the sugen chronic hypoxia model rat. We show that acute hexarelin administration evokes coronary microvascular dilation through GHS-receptor 1a and nitric oxide, and through endothelium-derived hyperpolarization. Previous work indicated that chronic exogenous administration of ghrelin largely prevented the pathogenesis of pulmonary hypertension in chronic hypoxia and in monocrotaline models. Unexpectedly, chronic hexarelin administration prior to sugen chronic hypoxia did not prevent RV hypertrophy or RV cardiomyocyte relaxation impairment. Small-angle X-ray scattering revealed that super relaxed myosin filaments contributed to diastolic dysfunction, and that length-dependent activation might contribute to sustained contractility of the RV. Thus, synchrotron-based imaging approaches can reveal novel insights into cardiac and coronary functions in vivo.
... Growth hormone secretagogues (GHS), a class of synthetic peptides stimulating growth hormone release through binding of the G-protein coupled receptor (GHS-R) have been demonstrated to have CV actions [20,21]. A number of studies have demonstrated the protective effects of the synthetic hexapeptide, hexarelin (HEX) in AMI, IR injury and cardiac fibrosis [20,[22][23][24]. ...
... Growth hormone secretagogues (GHS), a class of synthetic peptides stimulating growth hormone release through binding of the G-protein coupled receptor (GHS-R) have been demonstrated to have CV actions [20,21]. A number of studies have demonstrated the protective effects of the synthetic hexapeptide, hexarelin (HEX) in AMI, IR injury and cardiac fibrosis [20,[22][23][24]. There are accumulating studies that describe the application of GHS in CV disease clinical trials with promising results [25,26], however there remains a paucity of data detailing the mechanisms underlying these effects. ...
... There are accumulating studies that describe the application of GHS in CV disease clinical trials with promising results [25,26], however there remains a paucity of data detailing the mechanisms underlying these effects. In preclinical disease models, there is evidence supporting the indirect cardioprotective action of GHS through central and peripheral modulation of the ANS [20,21,24,27]. ...
Article
Full-text available
Acute myocardial ischemia and reperfusion injury (IRI) underly the detrimental effects of coronary heart disease on the myocardium. Despite the ongoing advances in reperfusion therapies, there remains a lack of effective therapeutic strategies for preventing IRI. Growth hormone secretagogues (GHS) have been demonstrated to improve cardiac function, attenuate inflammation and modulate the autonomic nervous system (ANS) in models of cardiovascular disease. Recently, we demonstrated a reduction in infarct size after administration of hexarelin (HEX), in a murine model of myocardial infarction. In the present study we employed a reperfused ischemic (IR) model, to determine whether HEX would continue to have a cardioprotective influence in a model of higher clinical relevance. Myocardial ischemia was induced by transient ligation of the left descending coronary artery (tLAD) in C57BL/6 J mice followed by HEX (0.3 mg/kg/day; n = 20) or vehicle (VEH) (n = 18) administration for 21 days, first administered immediately prior-to reperfusion. IR-injured and sham mice were subjected to high-field magnetic resonance imaging to assess left ventricular (LV) function, with HEX-treated mice demonstrating a significant improvement in LV function compared with VEH-treated mice. A significant decrease in interstitial collagen, TGF-β1 expression and myofibroblast differentiation was also seen in the HEX-treated mice after 21 days. HEX treatment shifted the ANS balance towards a parasympathetic predominance; combined with a significant decrease in cardiac troponin-I and TNF-α levels, these findings were suggestive of an anti-inflammatory action on the myocardium mediated via HEX. In this model of IR, HEX appeared to rebalance the deregulated ANS and activate vagal anti-inflammatory pathways to prevent adverse remodelling and LV dysfunction. There are limited interventions focusing on IRI that have been successful in improving clinical outcome in acute myocardial infarction (AMI) patients, this study provides compelling evidence towards the translational potential of HEX where all others have largely failed.
... In humans, hexarelin actions are not restricted to stimulating GH release; to illustrate, acute hexarelin administration markedly increased left ventricular function in (i) normal subjects, (ii) in patients with ischemic cardiomyopathy, as well as (iii) in patients with severe GH deficiency (20). Hexarelin significantly reduced indices of cardiac fibrosis in experimental models of myocardial infarction, likely through an underlying anti-inflammatory mechanism (12). The effects of subacute hexarelin treatments in rats (8) strongly suggested that hexarelin could interfere with the renin-angiotensin-aldosterone system (RAAS). ...
... We have observed that hexarelin reduced the number of immune cells in the BAL, in particular PMN, suggesting that it could partially inhibit the recruitment of PMN into the alveolar space. This is consistent with the anti-inflammatory and antifibrotic effects reported for hexarelin in the heart (12). In this study, we observed that hexarelin significantly reduced OH-proline levels, a measure of collagen deposition, in the lung treated with acid instillation. ...
Article
Full-text available
Introduction: Acute respiratory distress syndrome (ARDS) is an acute form of diffuse lung injury characterized by (i) an intense inflammatory response, (ii) increased pulmonary vascular permeability, and (iii) the loss of respiratory pulmonary tissue. In this article we explore the therapeutic potential of hexarelin, a synthetic hexapeptide growth hormone secretagogue (GHS), in an experimental model of ARDS. Hexarelin has anti-inflammatory properties and demonstrates cardiovascular-protective activities including the inhibition of cardiomyocyte apoptosis and cardiac fibrosis, both of which may involve the angiotensin-converting enzyme (ACE) system. Methods: In our experimental model, ARDS was induced by the instillation of 100 mM HCl into the right bronchus; these mice were treated with hexarelin (320 μg/kg, ip) before (Pre) or after (Post) HCl challenge, or with vehicle. Respiratory system compliance, blood gas analysis, and differential cell counts in a selective bronchoalveolar lavage (BAL) were determined 6 or 24 hours after HCl instillation. In an extended study, mice were observed for a subsequent 14 days in order to assess lung fibrosis. Results: Hexarelin induced a significant improvement in lung compliance and a reduction of the number of total immune cells in BAL 24 hours after HCl instillation, accompanied with a lower recruitment of neutrophils compared with the vehicle group. At day 14, hexarelin-treated mice presented with less pulmonary collagen deposition compared with vehicle-treated controls. Conclusions: Our data suggest that hexarelin can inhibit the early phase of the inflammatory response in a murine model of HCl-induced ARDS, thereby blunting lung remodeling processes and fibrotic development.
... Ischemia/reperfusion (IR) injury may be defined as damage of cells that results from a period of ischemia followed by reestablishment of blood supply at the ischemic site [133]. It is a significant cause of myocardial morbidity after an acute MI [134][135][136]. Ghrelin has been shown to be beneficial against I/R injury in numerous organs including the liver, kidneys, intestines, spinal cord, and pancreas in animal models [137][138][139][140][141]. In the heart, both ghrelin and its analogue hexarelin have been shown to exert cardioprotective effects after I/R injury [110,111]. ...
... This negative inotropic effect of hexarelin [221] was reduced by blocking nitric oxide (NO) synthesis [208,222]. In contrast, hexarelin did not show any significant effect on calcium transients and L-type Ca 2+ current (I Ca ), in isolated ventricular cells [134,209]. Taken together, these findings indicate that the effects of hexarelin may act via nitric oxide (NO) release and prostacyclin (PGI2) from the endocardial endothelium, rather than direct effects on cardiomyocytes [209]. Infusion of ghrelin was also attenuated by inhibition of K ca channels, suggesting that these channels may play critical role in ghrelin-induced vasodilation [223]. ...
Article
Full-text available
Ghrelin, a small peptide hormone (28 aa), secreted mainly by X/A-like cells of gastric mucosa, is also locally produced in cardiomyocytes. Being an orexigenic factor (appetite stimulant), it promotes release of growth hormone (GH) and exerts diverse physiological functions, viz. regulation of energy balance, glucose, and/or fat metabolism for body weight maintenance. Interestingly, administration of exogenous ghrelin significantly improves cardiac functions in CVD patients as well as experimental animal models of heart failure. Ghrelin ameliorates pathophysiological condition of the heart in myocardial infarction, cardiac hypertrophy, fibrosis, cachexia, and ischemia reperfusion injury. This peptide also exerts significant impact at the level of vasculature leading to lowering high blood pressure and reversal of endothelial dysfunction and atherosclerosis. However, the molecular mechanism of actions elucidating the healing effects of ghrelin on the cardiovascular system is still a matter of conjecture. Some experimental data indicate its beneficial effects via complex cellular cross talks between autonomic nervous system and cardiovascular cells, some other suggest more direct receptor–mediated molecular actions via autophagy or ionotropic regulation and interfering with apoptotic and inflammatory pathways of cardiomyocytes and vascular endothelial cells. Here, in this review, we summarise available recent data to encourage more research to find the missing links of unknown ghrelin receptor–mediated pathways as we see ghrelin as a future novel therapy in cardiovascular protection.
... For example, hexarelin has demonstrated cardioprotective effects in rodent models of cardiovascular dysfunction after daily subcutaneous administrations for up to 5 weeks (Ghigo et al., 1999;Locatelli et al., 1999). Cardiotropic activity has also been observed after acute administration of hexarelin to normal and growth hormone-deficient humans as well as during by-pass surgery (Bisi et al., 1999;Ghigo et al., 1999;Mao et al., 2014). In this perspective, evidence is provided to suggest that the pharmacologic modulation of CD36 response could attenuate the macrophage-driven hyper-inflammatory response observed in SARS-CoV-2-infected patients. ...
Article
Full-text available
The scientific and medical community faced an unprecedented global health hazard that led to nearly 7 million deaths attributable to the rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In spite of the development of efficient vaccines against SARS-CoV-2, many people remain at risk of developing severe symptoms as the virus continues to spread without beneficial patient therapy. The hyper-inflammatory response to SARS-CoV-2 infection progressing to acute respiratory distress syndrome remains an unmet medical need for improving patient care. The viral infection stimulates alveolar macrophages to adopt an inflammatory phenotype regulated, at least in part, by the cluster of differentiation 36 receptor (CD36) to produce unrestrained inflammatory cytokine secretions. We suggest herein that the modulation of the macrophage response using the synthetic CD36 ligand hexarelin offers potential as therapy for halting respiratory failure in SARS-CoV-2-infected patients.
... Accumulating evidence suggests the use of various drugs, including chemical agents, natural products and combined therapies, for the treatment of IRI-mediated renal diseases, such as AKI, CKD and AKI-to-CKD [17][18][19]. Hexarelin, also known as Examorelin, is a synthetic analog of growth hormone releasing peptide 6 (GHRP-6) that exhibits chemically stable and potent functional properties in stimulating growth hormone release [20]. Recently, novel therapeutic effects of Hexarelin, beyond growth hormone release, have been demonstrated in studies investigating cardiac and neural IRI [21]. ...
Article
Full-text available
Introduction Hexarelin exhibits significant protection against organ injury in models of ischemia/reperfusion (I/R)-induced injury (IRI). Nevertheless, the impact of Hexarelin on acute kidney injury (AKI) and its underlying mechanism remains unclear. In this study, we investigated the therapeutic potential of Hexarelin in I/R-induced AKI and elucidated its molecular mechanisms. Methods We assessed the protective effects of Hexarelin through both in vivo and in vitro experiments. In the I/R-induced AKI model, rats were pretreated with Hexarelin at 100 μg/kg/d for 7 days before being sacrificed 24 h post-IRI. Subsequently, kidney function, histology, and apoptosis were assessed. In vitro, hypoxia/reoxygenation (H/R)-induced HK-2 cell model was used to investigate the impact of Hexarelin on apoptosis in HK-2 cells. Then, we employed molecular docking using a pharmmapper server and autodock software to identify potential target proteins of Hexarelin. Results In this study, rats subjected to I/R developed severe kidney injury characterized by tubular necrosis, tubular dilatation, increased serum creatinine levels, and cell apoptosis. However, pretreatment with Hexarelin exhibited a protective effect by mitigating post-ischemic kidney pathological changes, improving renal function, and inhibiting apoptosis. This was achieved through the downregulation of conventional apoptosis-related genes, such as Caspase-3, Bax and Bad, and the upregulation of the anti-apoptotic protein Bcl-2. Consistent with the in vivo results, Hexarelin also reduced cell apoptosis in post-H/R HK-2 cells. Furthermore, our analysis using GSEA confirmed the essential role of the apoptosis pathway in I/R-induced AKI. Molecular docking revealed a strong binding affinity between Hexarelin and MDM2, suggesting the potential mechanism of Hexarelin’s anti-apoptosis effect at least partially through its interaction with MDM2, a well-known negative regulator of apoptosis-related protein that of p53. To validate these findings, we evaluated the relative expression of MDM2 and p53 in I/R-induced AKI with or without Hexarelin pre-administration and observed a significant suppression of MDM2 and p53 by Hexarelin in both in vivo and in vitro experiments. Conclusion Collectively, Hexarelin was identified as a promising medication in protecting apoptosis against I/R-induced AKI.
... Specifically, the pretreatment of mice with subcutaneous injections of EP80317 followed by experimentally elicited infarction showed reduced injury and dysfunction when compared to sham injections, while no effect of treatment was observed in CD36 null mice [34]. Likewise, in rat studies, hexarelin, a synthetic growth hormone-releasing peptide that binds to the growth hormone receptor but also to CD36 was found to protect the heart from ischemia/reperfusion injury [40,41]. Exenatide, a long half-life analog of glucagon-like peptide 1 (GLP-1) also improves post-ischemic cardiac function in rats [42]. ...
Article
Full-text available
Introduction: Disturbances in myocardial lipid metabolism are increasingly being recognized as drivers of the development and progression of heart disease. Therefore, there is a need for treatments that can directly target lipid metabolic defects in heart failure. The membrane-associated glycoprotein CD36 plays a pivotal role in governing myocardial lipid metabolism by mediating lipid signaling and facilitating the cellular uptake of long-chain fatty acids. Emerging evidence suggests that CD36 is a prominent target in the treatment of heart failure. Areas covered: This article provides an overview of the key role of CD36 for proper contractile functioning of the healthy heart, its implication in the development of cardiac disease (ischemia/reperfusion, cardiac hypertrophy, diabetic cardiomyopathy) and its application as target to normalize cardiac metabolism as part of so-called metabolic modulation therapy. Expert opinion: CD36 appears a promising and effective therapeutic target in the treatment of heart failure. Natural compounds and chemical agents known to alter the amount or subcellular distribution of CD36, or inhibit its functioning, should be evaluated for their potency to correct cardiac metabolism and cure heart disease.
... Hexarelin is a synthetic hexapeptide ligand of the GHS-R1a, which is chemically more stable and functionally more potent when compared with ghrelin [44]; consequently, these characteristics make hexarelin a promising alternative to ghrelin [45]. Ghrelin has been demonstrated to protect several cell types such as adipocytes [46], osteoblasts [47], cardiomyocytes and endothelial cells [48] by inhibiting apoptotic stimuli. ...
Article
Full-text available
Hexarelin, a synthetic hexapeptide, exerts cyto-protective effects at the mitochondrial level in cardiac and skeletal muscles, both in vitro and in vivo, may also have important neuroprotective bioactivities. This study examined the inhibitory effects of hexarelin on hydrogen peroxide (H2O2)-induced apoptosis in Neuro-2A cells. Neuro-2A cells were treated for 24 h with various concentrations of H2O2 or with the combination of H2O2 and hexarelin following which cell viability and nitrite (NO2−) release were measured. Cell morphology was also documented throughout and changes arising were quantified using Image J skeleton and fractal analysis procedures. Apoptotic responses were evaluated by Real-Time PCR (caspase-3, caspase-7, Bax, and Bcl-2 mRNA levels) and Western Blot (cleaved caspase-3, cleaved caspase-7, MAPK, and Akt). Our results indicate that hexarelin effectively antagonized H2O2-induced damage to Neuro-2A cells thereby (i) improving cell viability, (ii) reducing NO2− release and (iii) restoring normal morphologies. Hexarelin treatment also reduced mRNA levels of caspase-3 and its activation, and modulated mRNA levels of the BCL-2 family. Moreover, hexarelin inhibited MAPKs phosphorylation and increased p-Akt protein expression. In conclusion, our results demonstrate neuroprotective and anti-apoptotic effects of hexarelin, suggesting that new analogues could be developed for their neuroprotective effects.
... Hexarelin is an analog of ghrelin that shows cardioprotective effects both in vivo and in vitro (via GHS-R1a) [12,13]. Hexarelin is chemically more stable and functionally more potent when compared with ghrelin [14], which makes hexarelin a promising substitute for ghrelin in the clinical applications. Although the relationship between epilepsy and grelin is known for a while, and it is also well-documented that hexarelin is a potent and reliable agent which mimics ghrelin's biological functions, as far as know, there are only a few studies about the effect of hexarelin on epilepsy and there is none focusing on neuroprotection in PTZ-induced epilepsy model. ...
Article
Full-text available
Recent studies have demonstrated that ghrelin receptors have antiepileptic effects. The aim of this study was to investigate the effect of ghrelin receptor agonist hexarelin on pentylenetetrazole (PTZ)-induced seizures and post-seizure hippocampal damage. In our study, we used 42 male 230-250 g Wistar Albino rats. Animals were divided into seven groups as control, saline (PTZ; 1 ml/kg serum physiologic), positive control (5 mg/kg diazepam), 50 µg/kg, 100 µg/kg, 200 µg/kg and 400 µg/kg hexarelin. 30 min after drugs administration at the indicated doses, PTZ was administered 45 mg/kg to induce an epileptic seizure. The animals were observed for 30 min. Seizure stages (according to the Racine Scale) and first myoclonic jerk times (FMJ). 24 hours after PTZ injection, passive avoidance test was performed, and then brain tissues were removed. After the routine histological process, serial sections from brain tissues were stained with toluidine blue to determine neuronal damage. The hippocampal Cornu Ammonis CA1, CA3 and dentate gyrus regions were evaluated histopathologically. Statistical evaluation of the data was performed by oneway ANOVA, and multiple comparisons were determined by the Tukey test. Statistical significance was defined at p<0.05. Obtained data suggest that 200 µg/kg and 400 µg/kg hexarelin decreased seizure stages and increased FMJ compared to PTZ group (p<0,05). In addition, 200 µg/kg and 400 µg/kg hexarelin improved retention time in passive avoidance compared to PTZ group (p<0,05). Furthermore, 200 µg/kg and 400 µg/kg hexarelin reduced neuronal damage in hippocampal CA1, CA3, and DG regions compared to PTZ group (p<0,05). However, all these effects of hexarelin were not observed at 50 µg/kg and 100 µg/kg (p>0,05). In conclusion, we suggest that hexarelin has protective effects on epileptic seizures and neuronal damage after PTZ dose-dependently
Preprint
Full-text available
Hexarelin, a synthetic hexapeptide, protects cardiac and skeletal muscles by inhibiting apoptosis, both in vitro and in vivo . Moreover, evidence suggests that hexarelin could have important neuroprotective bioactivity. Oxidative stress and the generation of free radicals has been implicated in the etiologies of several neurodegenerative diseases, including amyotrophic lateral sclerosis, Parkinson’s disease, Alzheimer’s disease, Huntington’s disease and multiple sclerosis. In addition to direct oxidative stress, exogenous hydrogen peroxide (H 2 O 2 ) can penetrate biological membranes and enhance the formation of other reactive oxygen species. The aim of this study was to examine the inhibitory influence of hexarelin on H 2 O 2 -induced apoptosis in Neuro-2A cells, a mouse neuroblastoma cell line. Our results indicate that H 2 O 2 reduced the viability of Neuro-2A cells in a dose-related fashion. Furthermore, H 2 O 2 induced significant changes in the morphology of Neuro-2A cells, reflected in the formation of apoptotic cell bodies, and an increase of nitric oxide (NO) production. Hexarelin effectively antagonized H 2 O 2 oxidative damage to Neuro-2A cells as indicated by improved cell viability, normal morphology and reduced nitrite (NO 2 ⁻ ) release. Hexarelin treatment of Neuro-2A cells also reduced mRNA levels of caspases−3 and −7 and those of the pro-apoptotic molecule Bax; by contrast, hexarelin treatment increased anti-apoptotic Bcl-2 mRNA levels. Hexarelin also reduced MAPKs phosphorylation induced by H 2 O 2 and concurrently increased p-Akt protein expression. In conclusion, our results identify several neuroprotective and anti-apoptotic effects of hexarelin. These properties suggest that further investigation of hexarelin as a neuroprotective agent in an investigational and therapeutic context are merited.
Article
Full-text available
Both ghrelin and the synthetic analogue hexarelin are reported to possess cardioprotective actions that are mainly exerted through different receptors. However, their effects on acute myocardial infarction have not been compared in vivo. This study aimed to clarify whether hexarelin treatment can compensate for ghrelin deficiency in ghrelin knockout mice, and to compare the effects of hexarelin (400 nmol/kg per day, s.c.) and equimolar ghrelin treatment after myocardial infarction. Myocardial infarction was produced by left coronary artery ligation in male ghrelin-knockout mice, which then received ghrelin, hexarelin, or vehicle treatment for 2 weeks. The mortality within 2 weeks was significantly lower in the hexarelin group (6.7%) and ghrelin group (14.3%) than in the vehicle group (50%) (P < 0.05). A comparison of cardiac function 2 weeks post-infarction showed that in the ghrelin and hexarelin treatment groups, cardiac output were greater, while systolic function, represented by ejection fraction, and diastolic function, represented by dP/dt min, were significantly superior compared to the vehicle group (P < 0.05). Hexarelin treatment was more effective than ghrelin treatment, as indicated by the ejection fraction, dP/dt max, and dP/dt min. Telemetry recording and heart rate variability analysis demonstrated that sympathetic nervous activity was clearly suppressed in the hexarelin and ghrelin groups relative to the vehicle group. Our data demonstrated that hexarelin treatment can result in better heart function than ghrelin treatment 2 weeks after myocardial infarction in ghrelin knockout mice, although both hormones have similar effects on heart rate variability and mortality.
Article
Full-text available
Ischemic heart diseases often induce cardiac arrhythmia with irregular cardiac action potential (AP). This study aims to demonstrate that GH secretagogues (GHS) ghrelin and its synthetic analog hexarelin can preserve the electrophysiological properties of cardiomyocytes experiencing ischemia/reperfusion (I/R). Isolated hearts from adult male mice underwent 20 min global ischemia followed by 30 min reperfusion using a Langendorff apparatus. Ghrelin (10 nm) or hexarelin (1 nm) was administered in the perfusion solution either 10 min before or after ischemia, termed pre- or posttreatments. Cardiomyocytes isolated from these hearts were used for whole-cell patch clamping to measure AP, voltage-gated L-type calcium current (I(CaL)), transient outward potassium current (I(to)), and sodium current (I(Na)). AP amplitude and duration were significantly decreased by I/R, but GHS treatments maintained their normality. GHS treatments prevented the decrease in I(CaL) and I(Na) after I/R, thereby maintaining AP amplitude. Although the significant increase in I(to) after I/R partially explained the shortened AP duration, the normalization of it by GHS treatments might contribute to the preservation of AP duration. Phosphorylated p38 and c-Jun NH(2)-terminal kinase and the downstream active caspase-9 in the cellular apoptosis pathway were significantly increased after I/R but not when GHS treatments were included, whereas phosphorylation of ERK1/2 associated with cell survival showed increase after I/R and a further increase after GHS treatments by binding to its receptor GHS receptor type 1a. These results suggest GHS can not only preserve the electrophysiological properties of cardiomyocytes after I/R but also inhibit cardiomyocyte apoptosis and promote cell survival by modification of MAPK pathways through activating GHS receptor type 1a.
Article
Full-text available
Cardiac fibrosis is a hallmark of heart disease and plays a vital role in cardiac remodeling during heart diseases, including hypertensive heart disease. Hexarelin is one of a series of synthetic growth hormone secretagogues (GHSs) possessing a variety of cardiovascular effects via action on GHS receptors (GHS-Rs). However, the role of hexarelin in cardiac fibrosis in vivo has not yet been investigated. In the present study, spontaneously hypertensive rats (SHRs) were treated with hexarelin alone or in combination with a GHS-R antagonist for 5 wk from an age of 16 wk. Hexarelin treatment significantly reduced cardiac fibrosis in SHRs by decreasing interstitial and perivascular myocardial collagen deposition and myocardial hydroxyproline content and reducing mRNA and protein expression of collagen I and III in SHR hearts. Hexarelin treatment also increased matrix metalloproteinase (MMP)-2 and MMP-9 activities and decreased myocardial mRNA expression of tissue inhibitor of metalloproteinase (TIMP)-1 in SHRs. In addition, hexarelin treatment significantly attenuated left ventricular (LV) hypertrophy, LV diastolic dysfunction, and high blood pressure in SHRs. The effect of hexarelin on cardiac fibrosis, blood pressure, and cardiac function was mediated by its receptor, GHS-R, since a selective GHS-R antagonist abolished these effects and expression of GHS-Rs was upregulated by hexarelin treatment. In summary, our data demonstrate that hexarelin reduces cardiac fibrosis in SHRs, perhaps by decreasing collagen synthesis and accelerating collagen degradation via regulation of MMPs/TIMP. Hexarelin-reduced systolic blood pressure may also contribute to this reduced cardiac fibrosis in SHRs. The present findings provided novel insights and underscore the therapeutic potential of hexarelin as an antifibrotic agent for the treatment of cardiac fibrosis.
Article
Full-text available
Ischemic heart disease is a leading cause of mortality. To study this disease, ischemia/reperfusion (I/R) models are widely used to mimic the process of transient blockage and subsequent recovery of cardiac coronary blood supply. We aimed to determine whether the presence of the growth hormone secretagogues, ghrelin and hexarelin, would protect/improve the function of heart from I/R injury and to examine the underlying mechanisms. Isolated hearts from adult male mice underwent 20 min global ischemia and 30 min reperfusion using a Langendorff apparatus. Ghrelin (10 nM) or hexarelin (1 nM) was introduced into the perfusion system either 10 min before or after ischemia, termed pre- and post-treatments. In freshly isolated cardiomyocytes from these hearts, single cell shortening, intracellular calcium ([Ca(2+)](i)) transients and caffeine-releasable sarcoplasmic reticulum (SR) Ca(2+) were measured. In addition, RT-PCR and Western blots were used to examine the expression level of GHS receptor type 1a (GHS-R1a), and phosphorylated phospholamban (p-PLB), respectively. Ghrelin and hexarelin pre- or post-treatments prevented the significant reduction in the cell shortening, [Ca(2+)](i) transient amplitude and caffeine-releasable SR Ca(2+) content after I/R through recovery of p-PLB. GHS-R1a antagonists, [D-Lys3]-GHRP-6 (200 nM) and BIM28163 (100 nM), completely blocked the effects of GHS on both cell shortening and [Ca(2+)](i) transients. Through activation of GHS-R1a, ghrelin and hexarelin produced a positive inotropic effect on ischemic cardiomyocytes and protected them from I/R injury probably by protecting or recovering p-PLB (and therefore SR Ca(2+) content) to allow the maintenance or recovery of normal cardiac contractility. These observations provide supporting evidence for the potential therapeutic application of ghrelin and hexarelin in patients with cardiac I/R injury.
Article
We previously reported that induction of selective GH deficiency in the rat exacerbates cardiac dysfunction induced by experimental ischemia and reperfusion performed on the explanted heart. In the same model, short-term treatment with hexarelin, a GH-releasing peptide, reverted this effect, as did GH. To ascertain whether hexarelin had non-GH-mediated protective effects on the heart, we compared hexarelin and GH treatment in hypophysectomized rats. Hexarelin (80 μg/kg sc), given for 7 days, prevented exacerbation of the ischemia-reperfusion damage induced by hypophysectomy. We also demonstrate that hexarelin prevents increases in left ventricular end diastolic pressure, coronary perfusion pressure, reactivity of the coronary vasculature to angiotensin II, and release of creatine kinase in the heart perfusate. Moreover, hexarelin prevents the fall in prostacyclin release and enhances recovery of contractility. Treatment with GH (400 μg/kg sc) produced similar results, whereas administration of EP 51389 (8...
Article
Background: We have previously demonstrated the protective role of endogenous ghrelin against malignant arrhythmias in the very acute phase of myocardial infarction (MI). However, the role of endogenous ghrelin in the chronic phase is unknown. Therefore, the aim of the current study was to focus on the effects of endogenous ghrelin on cardiac function and sympathetic activation after acute MI. Methods and results: In 46 ghrelin-knockout (KO) and 41 wild-type (WT) male mice, MI was produced by left coronary artery ligation. The mortality due to heart failure within 2 weeks was 0% in WT and 10.9% in KO (P < 0.05). At the end of this period, lung weight/tibial length, atrial natriuretic peptide and brain natriuretic peptide transcripts, end-systolic and end-diastolic volumes were all significantly greater in KO mice, whereas systolic function, represented by ejection fraction (16.4 ± 4.7% vs. 25.3 ± 5.1%), end-systolic elastance, and preload-recruitable stroke work, was significantly inferior to that in WT mice (P < 0.05). Telemetry recording and heart rate variability analysis showed that KO mice had stronger sympathetic activation after MI than did WT mice. Metoprolol treatment and ghrelin treatment in KO mice prevented excessive sympathetic activation, decreased plasma epinephrine and norepinephrine levels, and improved heart function and survival rate after MI. Conclusions: Our data demonstrate that endogenous ghrelin plays a crucial role in protecting heart function and reducing mortality after myocardial infarction, and that these effects seem to be partly the result of sympathetic inhibition.
Article
Growth hormone releasing peptides (GHRPs) are synthetic molecules endowed with potent neuroendocrine activities mediated by specific receptors in the pituitary and in the central nervous system. GHRPs receptors have been reported even in perpheral tissues, particularly in the myocardium, where they probably mediate growth hormone (GH)-independent activities. We studied in humans the cardiac effects of hexarelin administration in 7 normal adults, in 7 severe GH-deficient patients, and in 12 patients with severe dilated cardiomyopathy. Left ventricular ejection fraction (LVEF), mean blood pressure (MBP), heart rate (HR), and GH levels were evaluated at baseline and every 15 min up to 60 min after acute 2.0 μg/kg iv hexarelin administration. Basal LVEF in dilated cardiomyopathy was impaired and lower (p<0.001) than in GH deficiency, in turn lower (p<0.001) than in normal subjects. Hexarelin significantly (p<0.05) increased LVEF in normal and in GH-deficient subjects, but not in dilated cardiomyopathy, without significant variations in MBP and HR. Hexeralin significantly (p<0.05) increased GH levels in normal subjects and in dilated cardiomyopathy but not in GH deficiency. These findings suggest that, in humans, the acute administration of hexarelin exerts a GH-independent positive inotropic effect likely mediated by specific GHRPs myocardial receptors.
Article
Ghrelin is a GH-releasing peptide mainly excreted from the stomach. Ghrelin administration has been shown to inhibit cardiac sympathetic nerve activity (CSNA), reduce malignant arrhythmia, and improve prognosis after acute myocardial infarction (MI). We therefore investigated the effects and potential mechanisms of the action of endogenous ghrelin on survival rate and CSNA after MI by using ghrelin-knockout (KO) mice. MI was induced by left coronary artery ligation in 46 KO mice and 41 wild-type mice. On the first day, malignant arrhythmia-induced mortality was observed within 30 min of the ligation and had an incidence of 2.4% in wild-type and 17.4% in KO mice (P < 0.05). We next evaluated CSNA by spectral analysis of heart rate variability. CSNA, represented by the low frequency/high frequency ratio, was higher in KO mice at baseline (2.18 ± 0.43 vs. 0.98 ± 0.09; P < 0.05), and especially after MI (25.5 ± 11.8 vs. 1.4 ± 0.3; P < 0.05), than in wild-type mice. Ghrelin (150 μg/kg, s.c.) 15 min before ligation suppressed the activation of CSNA and reduced mortality in KO mice. Further, this effect of ghrelin was inhibited by methylatropine bromide (1 mg/kg, i.p.) or by perineural treatment of both cervical vagal trunks with capsaicin (a specific afferent neurotoxin). Our data demonstrated that both exogenous and endogenous ghrelin suppressed CSNA, prevented the incidence of malignant arrhythmia, and improved the prognosis after acute MI. These effects are likely to be via the vagal afferent nerves.
Article
Ghrelin is a 28-amino acid peptide hormone produced in the stomach. It binds to the growth hormone secretagogue receptor 1a (GHS-R1a), a class A G-protein-coupled receptor. In the present study, we describe the design, synthesis and characterization of a truncated, 18-amino acid analog of ghrelin conjugated to a fluorescent molecule, fluorocein isothiocyanate (FITC), through the addition of a lysine at its C terminus ([Dpr(octanoyl)(3), Lys(fluorescein)(19)]ghrelin(1-19)). Receptor binding affinity of this novel fluorescein-ghrelin(1-18) was similar to that of wild-type ghrelin and a synthetic GHS-R1a ligand, hexarelin. Live cell imaging in CHO/GHS-R1a cells demonstrated cell surface receptor labeling and internalization, and agonist activity of fluorescein-ghrelin(1-18) was confirmed by increased phosphorylation of ERK1/2. We also show that GHS-R1a protein is expressed primarily in the heart when compared to all other organs, suggesting high receptor density in the left ventricle. Finally, we demonstrate that fluorescein-ghrelin(1-18) binds specifically to heart tissue in situ, and its binding is displaced by both wt ghrelin and hexarelin. We have therefore developed a novel imaging probe, fluorescein-ghrelin(1-18), that can be used to image GHS-R1a in situ, for the purposes of investigating mechanisms of receptor trafficking or pharmacological agents that target GHS-R1a.