Sie sind auf Seite 1von 11

Current Hypertension Reviews, 2011, 7, 273-283 273

Adrenomedullin in Heart Failure: Molecular Mechanism and Therapeutic


Implication

Toshio Nishikimi1,*, Kazuwa Nakao1 and Kenji Kangawa2

1
Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawara-
cho, Sakyo-ku, Kyoto 606-8507, Japan 2Department of Biochemistry, National Cerebral and Cardiovascular Center
Research Institute, Fujishirodai, Suita, Osaka 565-8565, Japan

Abstract: Many neurohumoral factors play an important role in regulation of the cardiovascular system and in the
pathophysiology of heart failure. Adrenomedullin (AM) is a potent long-lasting vasodilatory peptide that was discovered
in acid extracts of human pheochromocytoma tissues. Both AM and its gene expression are widely distributed in the
cardiovascular system, including the heart, vessels, and kidneys. AM co-localizes with its receptor components such as
calcitonin receptor-like receptor (CRLR), receptor activity modifying protein (RAMP)2 and RAMP3 in the heart, vessels
and kidneys, suggesting that it plays an important role in the regulation of cardiovascular function through an autocrine
and/or paracrine mechanism. AM has inotropic action in vitro and in vivo and inhibits cardiac hypertrophy in myocytes as
well as proliferation and collagen production in cardiac fibroblasts. These results suggest that AM functions as an
antifibrotic, antihypertrophic and positive inotropic factor in the failing heart. In addition, AM has anti-apoptotic,
angiogenic, anti-inflammatory and anti-oxidant effects. Several mechanisms such as cAMP/PKA, NO/cGMP, PI-3K/Akt
and/or ERK are thought to mediate cellular AM signaling, suggesting that increased AM levels play a protective role in
heart failure. Indeed, acute AM administration exerts beneficial vasodilatory, diuretic, natriuretic and inotropic effects on
experimental and human heart failure. A recent pilot study has shown that AM has potential as a therapeutic drug in the
clinical setting of acute decompensated heart failure.

Keywords: Adrenomedullin, heart failure, antifibrotic effects, cardiovascular diseases.

INTRODUCTION actions related to regulation of the cardiovascular system


have been identified [5]. The cardiovascular system, including
Heart failure has become an increasingly important
the heart, kidneys and vessels, expresses considerable
public health problem, because heart failure is the leading
amounts of AM mRNA [6] and immunoreactive AM is also
cause of hospitalization and death for people aged 65 years
widely distributed in these organs. These findings suggest
and over in the developed countries. Various neurohumoral
that AM plays a role in cardiovascular function and/or the
factors are involved in the pathophysiology of heart failure
pathophysiology of cardiovascular disease, and thus, this
[1]. Therefore, the pathophysiological roles of novel
protein has attracted considerable interest among cardio-
neurohumoral factors in heart failure should be investigated,
vascular investigators. We describe recent advances in the
since the findings can lead to the development of new drugs
biochemistry, receptor, signaling and cardiovascular action
[2]. In fact, angiotensin-converting enzyme inhibitors, beta-
of AM, its pathophysiological roles in the failing heart and
blockers, aldosterone blockers, and angiotensin receptor
clinical applications in heart failure.
blockers, which were generated in this manner, are now
widely used worldwide for patients with heart failure 1. STRUCTURE AND TISSUE DISTRIBUTION OF AM
and they have significantly contributed to improvements
Human AM consists of 52 amino acids, of which the
in prognoses and the quality of life for patients with this
carboxy-terminal tyrosine (Tyr) is amidated (Fig. 1), and one
syndrome.
intramolecular disulfide bond [3]. The sequence homology
Adrenomedullin (AM) is a powerful vasodilatory peptide of AM with human calcitonin gene-related peptide (CGRP)
that was discovered in extracts of human pheochromocytoma and amylin is only about 30%, even though each of these
tissues in 1993 by monitoring cAMP activity in rat platelets peptides has carboxy-terminal amides and a six-residue ring
[3]. Subsequent studies revealed various biological actions in structure formed by an intramolecular disulfide linkage. A
addition to its vasodilatory activity [4]. Specifically, many study of structure-activity relationships has shown that
amidation of the C-terminal residue of AM and its cyclic
structure are essential for receptor binding and the cAMP
*Address correspondence to this author at the Department of Medicine and response [7]. AM is therefore considered a member of the
Clinical Science, Kyoto University Graduate School of Medicine, 54 Shogoin- CGRP superfamily. The sequence identity of recently
Kawara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Tel: 81-75-751-4287; discovered AM 2 (AM2)/intermedin [8, 9] with AM is about
Fax: 81-75-771-9452; E-mail nishikim@kuhp.kyoto-u.ac.jp

1573-4021/11 $58.00+.00 © 2011 Bentham Science Publishers


274 Current Hypertension Reviews, 2011, Vol. 7, No. 4 Nishikimi et al.

Fig. (1). Biosynthesis of adrenomedullin (AM). Adrenomedullin, pro-AM N-terminal 20 peptide (PAMP) and mid-regional AM are
synthesized from the same precursor (prepro-AM: 185 amino acids). The signal peptide is removed and pro-AM is produced. Pro-AM is then
processed to glycine-extended AM (AM-glycine), glycine-extended PAMP and mid-regional AM. AM-glycine and glycine-extended PAMP
are inactive intermediate forms of AM and PAMP. Thereafter, AM-glycine and glycine-extended PAMP are converted to active mature AM
(AM-mature) and PAMP with a C-terminal amide structure by enzymatic amidation. Total AM (AM-T) = (AM-mature) + (AM-glycine).

30%, but the pharmacological activities (renal and cardio- showed that co-transfection of calcitonin like receptor (CLR)
vascular) of these peptides are similar [10]. Thus, at least and RAMP1 into COS-7 cells resulted in functional CGRP
two peptides comprise the AM family in mammals. receptors, whereas transfection of either gene alone elicited
no effects (Fig. 2) [16]. In addition, a database search
Immunoreactive AM is widely distributed in various
identified two more members of the RAMP family,
tissues. Levels of AM mRNA are notably high in
designated as RAMP2 and RAMP3 [15]. Although sequence
cardiovascular tissues such as ventricles, aorta, kidneys and
similarity among these RAMP isoforms is <30% within the
lungs, whereas levels of immunoreactive AM in the aorta,
same species, all three of them share conserved cysteine
ventricles and kidney are <5% of that in the adrenal gland
residues at their extracellular domain and flanking the
[11]. These low levels are attributed to the fact that AM
transmembrane domain [16]. Co-transfection of CRLR with
biosynthesized in these tissues is rapidly and constitutively
RAMP2 or RAMP3 leads to functional AM receptors with
secreted into the blood or utilized as an autocrine or
high affinity for AM, but not for CGRP (Fig. 2). In terms of
paracrine regulator, or both [12]. Immunohistochemical
AM binding and cAMP responses, RAMP2 and RAMP3 are
analyses have demonstrated AM immunoreactivity in cardiac
apparently indistinguishable [16]. The co-expression of
myocytes, vascular smooth muscle cells, endothelial cells
CRLR with RAMP1 and RAMP2/3 in fact results in
and renal distal and collecting tubules [13]. Immunoreactive
pharmacologic activities of CGRP1 and AM receptors,
AM is also found in blood and urine [14].
respectively (Fig. 2) [17].
2. RECEPTOR AND CELLULAR SIGNALS OF AM
Although AM was discovered based on elevated cAMP
McLatchie et al. [15] identified a cDNA encoding 148 activity in rat platelets, subsequent studies revealed many
amino-acid residues with a single transmembrane domain, cellular signaling mechanisms. AM receptors couple to Gq
named receptor activity modified protein (RAMP). They protein and AM activates phospholipase C to form inositol
Adrenomedullin in Heart Failure Current Hypertension Reviews, 2011, Vol. 7, No. 4 275

Fig. (2). Adrenomedullin receptor, signaling, downstream cascade and actions. Calcitonin receptor-like receptor pathway (CLR) and
receptor activity modifying protein (RAMP) isoforms determine ligand selectivity for AM and calcitonin-gene related peptides (CGRP).
Extracellular RAMP domains play important roles in ligand selectivity.

1,4,5-trisphosphate, and then activates nitric oxide synthase kinase (ERK) activity, followed by the expression of
in endothelial cells [18]. A later study showed that AM the immediate early proto-oncogene c-fos and promotes
induces Akt activation via the Ca/calmodulin-dependent proliferation in quiescent vascular smooth muscle cells [23].
pathway, leading to NO production [19]. The AM-induced AM also induces ERK activation in adipocytes [24], but
phosphatidylinositol 3'-kinase/Akt pathway plays a protective decreases ERK activity in mesangial cells [25]. The
role against endothelial cell apoptosis [20]. Thus, the AM/ mechanism through which AM decreases ERK activity is
Akt/PI-3K pathway plays important roles in eNOS activation thought to be mediated at least in part by an increase in
and apoptosis. During angiogenesis, AM induces the protein phosphatase 2A activity in mesangial cells [25].
phosphorylation of Akt and of mitogen-activated protein
Cardiac AM induces cAMP activity in myocytes and
kinase (MAPK)/ERK in human umbilical vein endothelial
cardiac fibroblasts [26] and inhibits myocyte growth [27] as
cells (HUVECs). AM also phosphorylates focal adhesion
well as collagen production [28] in fibroblasts. However, the
kinase, and phosphatidylinositol 3'-kinase inhibitor inhibits
cAMP/PKA pathway does not mediate the inotropic action
AM-induced focal adhesion kinase phosphorylation [21].
of AM, which enhances cardiac contractility via cAMP-
Thus, AM exerts angiogenic activity through the activation
independent mechanisms including Ca2+ release from intra-
of Akt, MAPK, and focal adhesion kinase in endothelial
cellular ryanodine- and thapsigargin-sensitive Ca2+ stores,
cells.
protein kinase C activation and Ca2+ influx through L-
AM induces cAMP in vascular smooth muscle cells, type Ca2+ channels [29]. In addition, AM increases the
which leads to vasodilation and the inhibition of proliferation phosphorylation of Akt and glycogen synthase kinase
and migration [22]. In contrast, another study showed that (GSK-3beta) and reduces caspase-3 activities in the heart.
AM induces a rapid increase in extracellular signal-regulated AM protects against cardiomyocyte apoptosis induced by
276 Current Hypertension Reviews, 2011, Vol. 7, No. 4 Nishikimi et al.

ischemia/reperfusion injury through the Akt-GSK-caspase pathway that decreases cytoplasmic Ca2+ concentrations. In
signaling pathway [30]. Thus, AM signaling processes are contrast, AM is reported to have direct inotropic activity in
complex and different mechanisms seem to be involved vitro [29] and increases myocardial contractility in vivo [35].
depending on the cellular situation. Szokodi et al. [29] demonstrated that AM has positive
inotropic action via cAMP-independent mechanisms and
3. PHYSIOLOGICAL EFFECTS OF AM
showed that AM is an endogenous inotropic peptide in the
Table 1 shows representative pleiotropic effects of AM. heart. The same group examined whether AM can modulate
the increase in blood pressure and changes in systolic and
Table 1. Physiological Action of Adrenomedullin diastolic function produced by long-term therapy with
angiotensin II or norepinephrine in rats. They found that AM
selectively suppresses the increase in blood pressure and
1. Vessel
improves the systolic function induced by angiotensin II,
Vasodilatation suggesting that circulating AM mainly acts as a regulator of
Nitric oxide production vascular tone and cardiac function [36]. In agreement with
Angiogenesis these findings, intravenous AM enhances left ventricular
2. Heart myocardial contraction and improves left ventricular
relaxation without increasing myocardial oxygen consump-
Inotropic action
tion in patients with old myocardial infarctions and left
3. Kidney ventricular dysfunction [37].
Glomerulus
AM knockout mice were first generated in 2001 [38].
Dilatation of afferent arteriole and constriction of efferent arteriole The phenotype is severe edema and cardiac abnormalities
Renal Tubule that lead to embryonic lethality [39]. Aortic constriction
Diuresis reduces ejection fraction more in AM+/- than in wild type
Natriuresis mice [39]. These results suggest that the AM/CLR/RAMP2
system preserves cardiac function as an endogenous factor.
4. Endocrine organ
Inhibition of aldosterone secretion The reasons for the discrepancies among studies
are unknown, but may involve differences in species,
Inhibition of ACTH secretion
experimental conditions, or the ages of the animals. The
5. Cell Growth, Proliferation, Apoptosis results of animal studies in vivo seem to consistently indicate
Inhibition of proliferation in vascular smooth muscle cells, that AM has inotropic action. Since it has inotropic activity
mesangial cells, and cardiac fibroblasts, in humans [37, 40], AM might be clinically applicable to
Growth-promoting effect in vascular smooth muscle cells patients with acute heart failure.
Inhibition of hypertrophy in cardiac myocytes
3-1-2. Cardiac Actions of AM - Effects on Cardiac
Antiapototic effect
Hypertrophy
Anti-inflammatory effects
AM significantly reduces 14C-phenylalanine incorporation
6. Adipocyte
into cardiac myocytes stimulated by 10% fetal bovine serum
lipolysis and angiotensin II [28]. This effect is dose-dependently
ACTH: adrenocorticotropic hormone. abolished by the AM receptor antagonist, CGRP[8-37].
AM also significantly and dose-dependently reduces the
angiotensin II- or ET-1-stimulated incorporation of 3H-
3-1-1. Cardiac Actions of AM - Effect of AM on Cardiac
thymidine and 3H-phenylalanine into cardiac fibroblasts [29,
Contractility
41], and CGRP[8-37] attenuates these effects. AM dose-
Many physiological actions of AM in the heart have been dependently stimulates cAMP accumulation in these cells
described [5]. The intravenous administration of AM in [26], an effect that is significantly attenuated by CGRP
animals significantly increases cardiac output [31], but [8-37]. Both 8-bromo cAMP and forskolin suppress the
whether this is a direct effect of AM or a secondary effect synthesis of DNA and collagen by cardiac fibroblasts.
resulting from reduced afterload remains unclear. An early Furthermore, a cAMP-specific phosphodiesterase inhibitor
study initially indicated direct negative inotropic actions of decreases the synthesis of both DNA and collagen in
AM in the perfused rat heart [32]. Others also reported that fibroblasts and augments the inhibitory effects of AM on
AM dose-dependently reduces both contractility and Ca2+ such synthesis [27]. AM antisense oligodeoxynucleotides
concentrations in isolated rabbit cardiac myocytes through significantly decrease AM levels in culture medium and
the NO/cGMP pathway [33]. Another group showed that collagen synthesis in cytokine-stimulated cardiac fibroblasts
AM augments NO synthesis in the heart through a cAMP [42], suggesting that endogenous AM indeed functions as an
pathway in response to cytokine stimulation [34]. These anti-fibrotic factor. These results suggest that AM has an
findings suggest that increased AM in the heart leads to important role in modulating the growth of cardiac
reduced cardiac contractility through a nitric oxide/cGMP fibroblasts in an autocrine or a paracrine fashion, at least in
Adrenomedullin in Heart Failure Current Hypertension Reviews, 2011, Vol. 7, No. 4 277

part via a cAMP signaling mechanism. The effect of AM on similarly dilates both afferent and efferent arterioles. They
cardiac hypertrophy was assessed in an in vivo rat model of also showed that the intrarenal arterial administration of AM
hypertrophy induced by aortic banding [43]. Both AM and induces an increase in urinary flow rates and urinary sodium
hydralazine similarly decreased blood pressure compared excretion even at a low dose without affecting glomerular
with an untreated group with aortic banding, but the left filtration rates. These findings suggest that AM inhibits
ventricular weight/body weight ratio was reduced only in the tubular sodium and water reabsorption. AM increases the
group infused with AM. Left ventricular AM levels were diameters of both afferent and efferent arterioles in the rat
22% greater and plasma AM levels were about 5-fold higher hydronephrotic kidney [52]. AM-induced renal vasodilator,
in the group infused with AM than in the untreated group diuretic and natriuretic responses might be partially mediated
with aortic banding. These findings have been supported by by the release of endogenous nitric oxide [53, 54] and renal
studies of engineered mice. AM heterozygous KO mice prostaglandins [55]. The inhibition of neutral endopeptidase
develop more pronounced fibrosis and left ventricular (NEP) potentiates an increase in sodium excretion in the
hypertrophy than wild type mice after aortic constriction absence of an increase in the glomerular filtration rate
[39]. The activation of ERK was more enhanced in the heart or further increases in renal blood flow in response to
after aortic constriction in AM heterozygous KO mice exogenous AM [56], indicating that NEP induces natriuresis
than in wild type mice. These findings suggest that AM by inhibiting tubular sodium reabsorption through attenuation
plays a pathophysiological role in the development of of AM metabolism. Collectively, these results suggest that
left ventricular hypertrophy. In addition, AM inhibits endogenous AM compensates for pathological conditions
cardiomyocyte apoptosis induced by doxorubicin [44] and such as atrial natriuretic peptide under specific pathological
ischemia/reperfusion injury [45]. Thus, AM may act as an conditions such as heart failure.
antifibrotic, antihypertrophic, antiapoptotic and positive
3-4. Endocrine Action of AM
inotropic factor in the failing or hypertrophied heart.
AM inhibits aldosterone secretion induced by angiotensin
3-2. Vascular Action of AM
II [57], potassium [58], and Ca2+ ionophores [59] in dispersed
The systemic intravenous injection of AM elicits a potent ZG cells and prevents increases in the plasma aldosterone
and long-lasting hypotensive effect in anesthetized rats [3] level in vivo induced by an infusion of angiotensin II [60], a
that is closely associated with a decrease in total peripheral sodium-deficient diet or bilateral nephrectomy [61]. These
resistance and concomitant with increases in cardiac output results suggest that AM plays an inhibitory role in the
and stroke volume [31]. In addition, acute or chronic AM secretion of aldosterone from zona glomerulosa cells and
administration induces a significant decrease in systemic that this inhibitory effect plays a role in pressure and volume
blood pressure in rabbits, sheep and humans. Vasodilatory homeostasis.
actions of AM have been studied in not only the systemic AM immunoreactivity has been detected in the
vasculature, but also in regional vascular beds, including the hypothalamo-pituitary-adrenal axis of humans, rats and
renal, pulmonary, cerebral and coronary circulation [23]. pigs [62], suggesting that AM functions in modulating the
These vasodilatory effects of AM are mediated by at least a secretion of pituitary and adrenal hormones. Indeed, AM
direct action on vascular smooth muscle cells and an indirect dose-dependently inhibits adrenocorticotropic hormone
effect through primary actions on endothelial cells via nitric (ACTH) release and attenuates CRH-stimulated ACTH
oxide production. production from primary cultures of rat anterior pituitary
3-3. Effect of AM on Renal Function cells [63]. This effect is also evident in vivo [64]. These
findings suggest that AM functions in inhibiting ACTH
AM has renal, as well as hypotensive actions. AM and its release. Thus, AM in the hypothalamo-pituitary complex
gene are expressed in the glomerulus, distal tubules and might play a role in the regulation of fluid and electrolyte
medullary collecting duct cells of the kidney [46, 47]. The homeostasis and modify the pathophysiology of heart failure.
genes encoding CRLR, RAMP2, and RAMP3, which are
components of AM receptors, are also expressed in the rat 4. PLASMA AM LEVELS
renal cortex and medulla [48]. Immunohistochemical analysis AM is found in plasma. Although high levels of AM
of the human kidney has revealed CRLR-like immuno- peptide and mRNA are expressed in the adrenal gland, heart,
reactivity in the juxtaglomerular arteries, the glomerular kidneys and lungs, this is not true of the coronary sinus,
capillaries and chief cells of the collecting duct [49]. These renal vein, adrenal veins and aorta [65]. Therefore, the
results suggest that AM and its receptor system in the kidney source of the plasma AM is now considered to be the
are involved in the regulation of renal hemodynamics, vasculature, because AM mRNA is more prominently
glomerular filtration and tubular sodium homeostasis in vivo. expressed in endothelial and vascular smooth muscle cells.
An intravenous infusion of AM attenuates a reduction in Plasma AM levels are increased in proportion to the severity
blood pressure and an increase in renal blood flow [50]. An of heart failure [66-69].
early study found that the intrarenal arterial administration of AM is produced from a precursor via two enzymatic
AM in the anesthetized dog elicits a dose-dependent increase steps. Firstly, the signal peptide [1-21] is removed from
in renal blood flow with a slight increase in the glomerular prepro-AM [1-185] to produce pro-AM [22-185], which is
filtration rate at high doses [51], suggesting that AM converted into glycine-extended AM (AM-Gly), glycine-
278 Current Hypertension Reviews, 2011, Vol. 7, No. 4 Nishikimi et al.

extended PAMP and mid-regional AM by a processing an increase in ventricular weight or fetal cardiac gene
enzyme. The inactive 53-amino-acid intermediate form of expression [76]. Plasma AM levels in human heart failure
AM, AM-Gly, is then converted by enzymatic amidation into are higher in coronary sinus than in the aorta, suggesting that
active mature AM (AM-m), a 52-amino-acid peptide with a cardiac AM production is increased in the failing heart.
C-terminal amide structure (Fig. 1). Both AM-Gly and AM-
Two molecular forms of AM, AM-mature and AM-
m are increased in heart failure [70]. A mid-regional stable
glycine (see Fig. 1), its mRNA, and receptors (CLR and
AM fragment of 45-92 amino acids that is secreted in an
RAMPs) have been investigated in cardiac hypertrophy and
equimolar fashion [71, 72] is an important prognostic marker
heart failure. The gene expression levels of AM, CLR,
of heart failure and myocardial infarction as well as BNP and
RAMP2, and RAMP3 in the left ventricle are significantly
NT-proBNP [73, 74].
higher in cardiac hypertrophy and in the failing heart than in
5. ROLE OF THE CARDIAC AM SYSTEM IN CAR- controls [77, 78]. Cardiac tissue levels of AM-mature and
DIAC HYPERTROPHY AND THE FAILING HEART AM-glycine were also significantly higher in cardiac
hypertrophy and in the failing heart than in controls, and the
AM immunoreactivity is increased in the failing heart
AM-mature/AM-glycine ratio is also significantly higher in
[67] and both mRNA and AM immunoreactivity are
left ventricular tissue than in plasma, suggesting the
increased in a model of heart failure [75]. In addition, the
increased amidating enzymatic activity. Furthermore, this
increased AM immunoreactivity in pressure-overloaded
AM-mature/AM-glycine ratio in left ventricular tissue
cardiac hypertrophy closely correlates with left ventricular
significantly correlates with the ratio of left ventricular
mass [43]. Thus, cardiac AM levels are upregulated in
weight/body weight [78, 79]. These results suggest that the
cardiac hypertrophy and the failing heart in association with
amidating enzymatic activity of AM, its ligand, and receptor

Fig. (3). Our working hypothesis regarding the myocardial and circulating AM system in transition from hypertension ~ LVH to
heart failure. In hypertension ~ LVH, plasma AM-mature and AM-glycine are increased. In myocardium, AM-glycine (inactive form) is
increased and AM-mature (active form) is further increased, because amidating enzyme activity (see Fig. 1) is increased in LVH. Receptor
mRNA levels are also increased. In heart failure, plasma, AM-mature and AM-glycine are further increased, whereas AM-mature/AM-
glycine ratio is not changed. In myocardium, AM-mature (active form) is markedly increased with a increased receptor mRNA levels. These
changes show the upregulation of AM system in plasma and myocardium in heart failure, compensating heart failure and inhibiting cardiac
remodeling. AM-m: AM-mature, AM-gly: AM-glycine, AM-m/gly ratio = AM-mature/AM-glycine ratio
Adrenomedullin in Heart Failure Current Hypertension Reviews, 2011, Vol. 7, No. 4 279

system are all upregulated in severe cardiac hypertrophy and induced heart failure [85]. AM also increases urinary
the failing heart. Thus, the entire cardiac AM system is sodium, creatinine and cAMP excretion, and creatinine
upregulated under these conditions (Fig. 3). clearance along with a reduction in plasma aldosterone
levels [85]. We also examined the cardiovascular and renal
Adenovirus-mediated AM gene delivery and the chronic
effects of two intravenous doses of infused AM in rats
administration of AM have been attempted to determine
with heart failure [86]. A low dose of AM increased urine
whether increased plasma and tissue AM levels in cardiac
flow and urinary sodium excretion without changing any
hypertrophy or the failing heart have cardioprotective effects.
hemodynamic variables, whereas a high dose decreased
Somatic gene delivery using an adenovirus containing human
mean arterial pressure, right ventricular systolic pressure and
AM cDNA under the control of the cytomegalovirus promoter/
right atrial pressure, and significantly increased cardiac
enhancer significantly decreased left ventricular weight
output in rats with heart failure and in normal rats. A high
and cardiomyocyte diameter, accompanied by reductions in
dose of AM significantly increased glomerular filtration rate
interstitial fibrosis, extracellular matrix formation and blood
and renal plasma flow, as well as urine flow and urinary
pressure in severely hypertensive rats [80]. These findings
sodium excretion [86]. In addition, AM administration
suggest that increased AM levels protect against cardiac
for four days has pronounced and sustained cardiovascular
remodeling and renal damage in hypertension.
and renal effects in experimental heart failure, including
The effects of long-term AM infusions have been reductions in cardiac preload and afterload as well as the
investigated in a rat model to determine the role of augmentation of cardiac output, sodium excretion and
endogenous AM in the transition from LVH to heart failure glomerular filtration [87]. These results imply that AM plays
[81]. Long-term infusions of human AM decreased left an important pathophysiological role in the regulation of
ventricular end-diastolic pressure, right ventricular systolic pressure and volume in heart failure and raises the prospect
pressure, right atrial pressure and the left ventricular of developing new therapeutic approaches to this syndrome.
weight/body weight ratio without significantly altering mean
AM, when combined with other drugs such as angiotensin
arterial pressure and significantly decreased endogenous
converting enzyme inhibitor, neutral endopeptidase inhibition
plasma AM levels in these model rats. In addition, long-
or natriuretic peptides, has beneficial and complementary
term AM infusion significantly decreased plasma renin,
effects [88-90].
aldosterone, and atrial natriuretic peptide levels and
prolonged survival. These results suggest that endogenous 7. EFFECTS OF AM IN PATIENTS WITH HEART
AM plays a compensatory role in heart failure. In addition FAILURE
to these salutary effects in heart failure, long-term AM The benefits determined in experimental heart failure
administration attenuates left ventricular remodeling after suggested that AM would be effective against human heart
acute myocardial infarction, partly via the inhibition of
failure.
oxidative stress and expression of the angiotensin converting
enzyme gene [82]. A protective effect of endogenous We found that an intravenous infusion of AM (0.05
AM against stress-induced cardiac hypertrophy has been μg/kg/min) for 30 minutes significantly increased the cardiac
investigated using the AM-null mutation AM+/- mice index and decreased pulmonary capillary wedge pressure in
described above [39]. patients with heart failure and in normal individuals [91].
AM significantly decreased mean pulmonary arterial
Thus, up-regulated cardiac expression of AM and its
pressure only in those with heart failure and increased urine
receptor might be an adaptive and protective response under
volume and urinary sodium excretion in both groups. Plasma
conditions of stress such as cardiac hypertrophy and heart
aldosterone fell significantly during and after AM infusion
failure. This hypothesis is supported by the results of recent
studies using AM gene delivery, long-term AM infusion only in the patients. These findings indicated that the acute
and knockout mice. The beneficial effects of increased AM intravenous infusion of AM has beneficial hemodynamic,
might be associated with the inhibition of oxidative stress, renal and endocrine effects in patients with heart failure [91].
the renin-angiotensin-aldosterone system and other factors The Christchurch group also studied the effects of AM
[22, 45, 81-84]. infusion on human heart failure [92] and found a significant
decrease in mean arterial pressure (MAP) and LV end-
6. ADMINISTRATION OF AM IN OTHER ANIMALS systolic volume, and increased cardiac output. Despite a
WITH HEART FAILURE large drop in MAP, urine volume, urinary sodium excretion
The above findings indicate that increased endogenous and creatinine clearance were not altered. These results
plasma and tissue levels of AM in cardiac hypertrophy and suggest that a short-term AM infusion improves heart failure
heart failure have cardioprotective effects. Together with and that AM can be therapeutically administered to patients
evidence of vasodilatory, diuretic, and natriuretic effects and with heart failure.
inhibited aldosterone secretion, AM might be a useful We then examined whether long-term AM (0.02
treatment strategy for patients with heart failure. μg/kg/min) + human atrial natriuretic peptide (hANP; 0.05
The intravenous infusion of AM decreases calculated μg/kg/min) can be administered as a therapeutic strategy to
peripheral resistance, mean arterial pressure and left atrial patients with acute decompensated heart failure (ADHF)
pressure, and increased cardiac output in sheep with pacing- in the clinical setting [93]. Seven patients with acute
280 Current Hypertension Reviews, 2011, Vol. 7, No. 4 Nishikimi et al.

Fig. (4). Effects of long-term administration of AM on hemodynamics in patients with decompensated heart failure. Effects of
AM + hANP and hANP monotherapy on mean arterial pressure (MAP), systemic vascular resistance (SVR), mean pulmonary arterial
pressure (mPAP), pulmonary arterial resistance (PAR), pulmonary capillary wedge pressure (PCWP) and cardiac index (CI) in patients with
decompensated heart failure. * P < 0.05 vs. time 0.

decompensated heart failure accompanied by dyspnea and CONCLUSIONS


pulmonary congestion were infused with AM and
Current evidence suggests that increased endogenous
recombinant ANP for 12 h followed by hANP for 12 h and
levels of AM play a protective role in cardiac hypertrophy
then hemodynamic, renal, hormonal and oxidative stress
and the failing heart. Thus, AM might be a novel and
responses were evaluated. AM + hANP significantly reduced
promising approach to the treatment of patients with heart
mean arterial pressure, pulmonary arterial pressure, and
failure. A small pilot study recently indicated that AM exerts
systemic and pulmonary vascular resistance without
beneficial therapeutic effects against ADHF. A larger
changing heart rate, and increased cardiac output at most
clinical trial is required to confirm this finding.
time points compared with baseline values (Fig. 4). This
combination also reduced aldosterone, BNP and free radical DISCLOSURE
metabolites, and increased urine volume and urinary sodium
Part of information included in this chapter/article has
excretion compared with baseline data. After switching to
been previously published in " http://www.ingentaconnect.
hANP monotherapy, MAP and systemic vascular resistance
com/content/ben/chyr Current Hypertension Reviews, Volume
increased and cardiac index decreased, but urinary volume
and urinary sodium excretion did not significantly change 1, Number 2, June 2005, pp. 169-181(13).
[93]. Although this was a small pilot trial, the combination of CONFLICT OF INTEREST
AM with hANP exerted beneficial hemodynamic and
hormonal effects in patients with ADHF. The ability of AM Declared none.
to reduce systemic and pulmonary vascular resistance and ACKNOWLEDGMENTS
increase the cardiac index was particularly prominent. Thus,
intravenous infusions of AM with hANP should be This study was supported in part by a Scientific Research
therapeutically applicable to patients with ADHF. However, Grant-in-Aid (14570692,18590787, 20590837) from the
these data are preliminary and require confirmation in a Ministry of Education, Culture, Sports, Science and
larger clinical study. Technology, by a Science Research Promotion Fund from
Adrenomedullin in Heart Failure Current Hypertension Reviews, 2011, Vol. 7, No. 4 281

the Promotion and Mutual Aid Corporation for Private [19] Nishimatsu H, Suzuki E, Nagata D, et al. Adrenomedullin induces
Schools of Japan, and by Suzuken memorial foundation. endothelium-dependent vasorelaxation via the phosphatidylinositol
3-kinase/Akt-dependent pathway in rat aorta. Circ Res 2001; 89:
We thank Dr. Naoto Minamino, Dr. Toshihiko Ishimitsu, 63-70.
[20] Kim W, Moon SO, Sung MJ, et al. Protective effect of
Dr. Takeshi Horio, Dr. Fumiki Yoshihara, Dr. Noritoshi adrenomedullin in mannitol-induced apoptosis. Apoptosis 2002; 7:
Nagaya, Dr. Koichiro Kuwahara, Dr. Yasuaki Nakagawa, 527-36.
and Dr. Hideyuki Kinoshita for helpful advice. We also [21] Kim W, Moon SO, Sung MJ, et al. Angiogenic role of adrenomedullin
thank Mr. Kazumim Akimoto, Mr. Hisato Hirata, Mr. through activation of Akt, mitogen-activated protein kinase, and
focal adhesion kinase in endothelial cells. FASEB J 2003; 17:
Yoshifumi Machida, Ms. Yasuko Mamada, Ms. Keiko 1937-9.
Fukuda, Ms. Kyoko Tabei, Ms. Masako Minato, and Ms. [22] Nishikimi T, Matsuoka H. Adrenomedullin in hypertension.
Machiko Sakata for excellent technical assistance. Current Hypertension Review 2005; 1: 169-181
[23] Iwasaki H, Eguchi S, Shichiri M, Marumo F, Hirata Y.
REFERENCES Adrenomedullin as a novel growth-promoting factor for cultured
vascular smooth muscle cells: role of tyrosine kinase-mediated
[1] Lee DS, Gona P, Albano I, et al. A systematic assessment of causes
mitogen-activated protein kinase activation. Endocrinology 1998;
of death after heart failure onset in the community: impact of age at
139: 3432-41.
death, time period, and left ventricular systolic dysfunction. Circ
[24] Iemura-Inaba C, Nishikimi T, Akimoto K, Yoshihara F, Minamino
Heart Fail 2011; 4: 36-43.
N, Matsuoka H. Role of adrenomedullin system in lipid
[2] Schrier RW, Abraham WT. Hormones and hemodynamics in heart
metabolism and its signaling mechanism in cultured adipocytes.
failure. N Engl J Med 1999; 341: 577-85.
Am J Physiol Regul Integr Comp Physiol 2008; 295: R1376-84.
[3] Kitamura K, Kangawa K, Kawamoto M, et al. Adrenomedullin: a
[25] Parameswaran N, Nambi P, Hall CS, Brooks DP, Spielman WS.
novel hypotensive peptide isolated from human pheochromocytoma.
Adrenomedullin decreases extracellular signal-regulated kinase
Biochem Biophys Res Commun 1993; 192: 553-60.
activity through an increase in protein phosphatase-2A activity in
[4] Samson WK. Adrenomedullin and the control of fluid and
mesangial cells. Eur J Pharmacol 2000; 388: 133-8.
electrolyte homeostasis. Annu Rev Physiol 1999; 61: 363-69.
[26] Nishikimi T, Horio T, Yoshihara F, Nagaya N, Matsuo H,
[5] Nishikimi T, Matsuoka H. Cardiac adrenomedullin: its role in
Kangawa K. Effect of adrenomedullin on cAMP and cGMP levels
cardiac hypertrophy and heart failure. Curr Med Chem Cardiovasc
in rat cardiac myocytes and nonmyocytes. Eur J Pharmacol 1998;
Hematol Agents 2005; 3: 231-42.
353: 337-44.
[6] Kitamura K, Sakata J, Kangawa K, Kojima M, Matsuo H, Eto T.
[27] Tsuruda T, Kato J, Kitamura K, et al. Adrenomedullin: a possible
Cloning and characterization of cDNA encoding a precursor for human
autocrine or paracrine inhibitor of hypertrophy of cardiomyocytes.
adrenomedullin. Biochem Biophys Res Commun 1993; 194: 720-5.
Hypertension 1998; 31: 505-10.
[7] Eguchi S, Hirata Y, Iwasaki H, et al. Structure-activity relationship
[28] Horio T, Nishikimi T, Yoshihara F, Matsuo H, Takishita S,
of adrenomedullin, a novel vasodilatory peptide, in cultured rat
Kangawa K. Effects of adrenomedullin on cultured rat cardiac
vascular smooth muscle cells. Endocrinology 1994; 135: 2454-8.
myocytes and fibroblasts. Eur J Pharmacol 1999; 382: 1-9.
[8] Roh J, Chang CL, Bhalla A, Klein C, Hsu SY. Intermedin is a
[29] Szokodi I, Kinnunen P, Tavi P, Weckstrom M, Toth M, Ruskoaho
calcitonin/calcitonin gene-related peptide family peptide acting
H. Evidence for cAMP-independent mechanisms mediating the
through the calcitonin receptor-like receptor/receptor activity-
effects of adrenomedullin, a new inotropic peptide. Circulation
modifying protein receptor complexes. J Biol Chem 2004: 279:
1998; 97: 1062-70.
7264-74.
[30] Yin H, Chao L, Chao J. Adrenomedullin protects against
[9] Takei Y, Inoue K, Ogoshi M, Kawahara T, Bannai H, Miyano S.
myocardial apoptosis after ischemia/reperfusion through activation
Identification of novel adrenomedullin in mammals: a potent
of Akt-GSK signaling. Hypertension 2004; 43: 109-16.
cardiovascular and renal regulator. FEBS Lett 2004; 56: 53-8.
[31] Ishiyama Y, Kitamura K, Ichiki Y, Nakamura S, Kida O, Kangawa
[10] Fujisawa Y, Nagai Y, Miyatake A, et al. Renal effects of a new
K, Eto T. Hemodynamic effects of a novel hypotensive peptide,
member of adrenomedullin family, adrenomedullin2, in rats. Eur J
human adrenomedullin, in rats. Eur J Pharmacol 1993; 241: 271-3.
Pharmacol 2004; 497: 75.
[32] Perret M, Broussard H, LeGros T, et al. The effect of
[11] Sakata J, Shimokubo T, Kitamura K, et al. Distribution and
adrenomedullin on the isolated heart. Life Sci 1993; 53: 377-9.
characterization of immunoreactive adrenomedullin in rat tissue
[33] Ikenouchi H, Kangawa K, Matsuo H, Hirata Y. Negative inotropic
and plasma. FEBS Lett 1994; 352: 105-8.
effect of adrenomedullin in isolated adult rabbit cardiac ventricular
[12] Bean AJ, Zhang X, Hoekfelt T. Peptide secretion: what do we
myocytes. Circulation 1997; 95: 2318-24.
know? FASEB J 1994; 8: 630-8.
[34] Ikeda U, Kanbe T, Kawahara Y, Yokoyama M, Shimada K.
[13] Washimine H, Asada Y. Kitamura K, et al. Immunohistochemical
Adrenomedullin augments inducible nitric oxide synthase
identification of adrenomedullin in human, rat, and porcine tissue.
expression in cytokine-stimulated cardiac myocytes. Circulation
Histochem Cell Biol 1995; 103: 251-4.
1996; 94: 2560-5.
[14] Sato K, Hirata Y, Imai T, Iwashina M, Marumo F. Characterization
[35] Parkes DG, May CN. Direct cardiac and vascular actions of
of immunoreactive adrenomedullin in human plasma and urine.
adrenomedullin in conscious sheep. Br J Pharmacol 1997; 120:
Life Sci 1995; 57: 189-94
1179-85.
[15] McLatchie LM, Fraser NJ, Main MJ, et al. RAMPs regulate the
[36] Luodonpaa M, Leskinen H, Ilves M, Vuolteenaho O, Ruskoaho H.
transport and ligand specificity of the calcitonin-receptor-like
Adrenomedullin modulates hemodynamic and cardiac effects of
receptor. Nature 1998; 393: 333-9.
angiotensin II in conscious rats. Am J Physiol Regul Integr Comp
[16] Born W, Muff R, Fischer JA. Functional interaction of G protein-
Physiol 2004; 286: R1085-92.
coupled receptors of the adrenomedullin peptide family with
[37] Nagaya N, Goto Y, Satoh T, et al. Intravenous adrenomedullin in
accessory receptor-activity-modifying proteins (RAMP). Microsc
myocardial function and energy metabolism in patients after
Res Tech 2002; 57: 14-22.
myocardial infarction. J Cardiovasc Pharmacol 2002; 39: 754-60.
[17] Poyner DR, Sexton PM, Marshall I, et al. International Union of
[38] Caron KM, Smithies O. Extreme hydrops fetalis and cardiovascular
Pharmacology. XXXII. The mammalian calcitonin gene-related
abnormalities in mice lacking a functional Adrenomedullin gene.
peptides, adrenomedullin, amylin, and calcitonin receptors.
Proc Natl Acad Sci USA 2001; 98: 615-9.
Pharmacol Rev 2002; 54: 233-46.
[39] Niu P, Shindo T, Iwata H, et al. Protective effects of endogenous
[18] Shimekake Y, Nagata K, Ohta S, et al. Adrenomedullin stimulates
adrenomedullin on cardiac hypertrophy, fibrosis, and renal damage.
two signal transduction pathways, cAMP accumulation and Ca2+
Circulation 2004; 109: 1789-94.
mobilization, in bovine aortic endothelial cells. J Biol Chem 1995;
270: 4412-7.
282 Current Hypertension Reviews, 2011, Vol. 7, No. 4 Nishikimi et al.

[40] Nishikimi T, Yoshihara F, Mori Y, Kangawa K, Matsuoka H. [63] Parkes DG, May CN. ACTH-suppressive and vasodilator actions of
Cardioprotective effect of adrenomedullin in heart failure. Hyp Res adrenomedullin in conscious sheep. J Neuroendocrinol 1995; 7:
2003; 26: S121-S127 923-9.
[41] Tsuruda T, Kato J, Kitamura K, et al. An autocrine or a paracrine [64] Nakamura Y, Shimatsu A, Murabe H, Mizuta H, Ihara C, Nakao K.
role of adrenomedullin in modulating cardiac fibroblast growth. Calcitonin gene-related peptide as a GH secretagogue in human
Cardiovasc Res 1999; 43: 958-67. and rat pituitary somatotrophs. Brain Res 1998; 807: 203-7.
[42] Nishikimi T, Tadokoro K, Akimoto K, et al. Response of [65] Nishikimi T, Kitamura K, Saito Y, et al. Clinical studies for the
adrenomedullin system to cytokine stimulation in cardiac sites of production and clearance of circulating adrenomedullin in
fibroblasts - Role of adrenomedullin as an antifibrotic factor- human subjects. Hypertension 1994; 24: 600-604
Cardiovasc Res 2005; 66: 94-103 [66] Nishikimi T, Saito Y, Kitamura K, et al. Increased plasma levels of
[43] Morimoto A, Nishikimi T, Yoshihara F, et al. Ventricular adrenomedullin in patients with heart failure. J Am Coll Cardiol
adrenomedullin levels correlate with the extent of cardiac 1995; 26: 1424-31.
hypertrophy in rats. Hypertension 1999; 33: 1146-52. [67] Jougasaki M, Wei CM, McKinley LJ, Burnett JC Jr. Elevation of
[44] Tokudome T, Horio T, Yoshihara F, et al. Adrenomedullin inhibits circulating and ventricular adrenomedullin in human congestive
doxorubicin-induced cultured rat cardiac myocyte apoptosis via a heart failure. Circulation 1995; 92: 286-9.
cAMP-dependent mechanism. Endocrinology 2002; 143: 3515-21. [68] Kato J, Kobayashi K, Etoh T, et al. Plasma adrenomedullim
[45] Kato K, Yin H, Agata J, Yoshida H, Chao L, Chao J. concentration in patients with heart failure. J Clin Endocrinol
Adrenomedullin gene delivery attenuates myocardial infarction and Metab 1996; 81: 180.
apoptosis after ischemia and reperfusion. Am J Physiol Heart Circ [69] Kobayashi K, Kitamura K, Etoh T, et al. Increased plasma
Physiol 2003; 285: H1506-14. adrenomedullin levels in chronic congestive heart failure. Am
[46] Asada Y, Hara S, Marutsuka K, et al. Novel distribution of Heart J 1996; 131: 1994-8.
adrenomedullin-immunoreactive cells in human tissues. Histochem [70] Hirayama N, Kitamura K, Imamura T, et al. Molecular forms of
Cell Biol 1999; 112: 185-91. circulating adrenomedullin in patients with congestive heart failure.
[47] Nishikimi T, Yoshihara F, Kanazawa A, et al. Role of increased J Endocrinol 1999; 160: 297-303
circulating and renal adrenomedullin in rats with malignant [71] Christ-Crain M, Morgenthaler NG, Struck J, Harbarth S,
hypertension. Am J Physiol Regul Integr Comp Physiol 2001; 281: Bergmann A, Miller B. Mid-regional pro-adrenomedullin as a
R2079-87. prognostic marker in sepsis: an observational study. Crit Care
[48] Yoshihara, F, Nishikimi T, Okano I, et al. Alterationsof intrarenal 2005; 9: R816-24.
adrenomedullin and its receptor system in heart failure rats. [72] Dhillon OS, Khan SQ, Narayan HK, et al. Prognostic value of mid-
Hypertension 2001; 37: 216. regional pro-adrenomedullin levels taken on admission and
[49] Hagner S, Stahl U, Knoblauch B, McGregor GP, Lang RE. discharge in non-ST-elevation myocardial infarction: the LAMP
Calcitonin receptor-like receptor: identification and distribution in (Leicester Acute Myocardial Infarction Peptide) II study. J Am
human peripheral tissues. Cell Tissue Res 2002; 310: 41-50. Coll Cardiol 2010; 56: 125-33.
[50] Vari RC, Adkins SD, Samson WK. Renal effects of adrenomedullin [73] Maisel A, Mueller C, Nowak R, et al. Mid-region pro-hormone
in the rat. Proc Soc Exp Biol Med 1996; 211: 178-83. markers for diagnosis and prognosis in acute dyspnea: results from
[51] Ebara T, Miura K, Okumura M, et al. Effect of adrenomedullin on the BACH (Biomarkers in Acute Heart Failure) trial. J Am Coll
renal hemodynamics and functions in dogs. Eur J Pharmacol 1994; Cardiol 2010; 55(19): 2062-76.
263: 69-73. [74] Klip IT, Voors AA, Anker SD, et al. OPTIMAAL investigators.
[52] Hirata Y, Hayakawa H, Suzuki Y, et al. Mechanisms of Prognostic value of mid-regional pro-adrenomedullin in patients
adrenomedullin-induced vasodilation in the rat kidney. Hypertension with heart failure after an acute myocardial infarction. Heart 2011;
1995; 25: 790-5. 97: 892-8.
[53] Majid DS, Kadowitz PJ, Coy DH, Navar LG. Renal responses to [75] Nishikimi T, Horio T, Sasaki T, et al. Cardiac production and
intra-arterial administration of adrenomedullin in dogs. Am J secretion of adrenomedullin are increased in heart failure.
Physiol 1996; 270: F200-5. Hypertension 1997; 30: 1369-75.
[54] Miura K, Ebara T, Okumura M, Matsuura T, Kim S, Yukimura T, [76] Yoshihara F, Nishikimi T, Horio T, et al. Ventricular
Iwao H. Attenuation of adrenomedullin-induced renal vasodilatation by adrenomedullin concentration is a sensitive biochemical marker for
NG-nitro L-arginine but not glibenclamide. Br J Pharmacol 1995; volume and pressure overload in rats. Am J Physiol Heart Circ
115: 917-24. Physiol 2000; 278: H633-42.
[55] Jougasaki M, Aarhus LL, Heublein DM, Sandberg SM, Burnett JC [77] Tadokoro K, Nishikimi T, Mori Y, Wang X, Akimoto K, Matsuoka
Jr. Role of prostaglandins and renal nerves in the renal actions of H. Altered gene expression of adrenomedullin and its receptor
adrenomedullin. Am J Physiol 1997; 272: F260-6. system and molecular forms of tissue adrenomedullin in left
[56] Lisy O, Jougasaki M, Schirger JA, Chen HH, Barclay PT, Burnett ventricular hypertrophy induced by malignant hypertension.Regul
JC Jr. Neutral endopeptidase inhibition potentiates the natriuretic Pept 2003; 112: 71-8.
actions of adrenomedullin. Am J Physiol 1998; 275: F410-4. [78] Nishikimi T, Tadokoro K, Mori Y, et al. Ventricular adrenomedullin
[57] Andreis PG, Mazzocchi G, Rebuffat P, Nussdorfer GG. Effects of system in the transition from LVH to heart failure in rats.
adrenomedullin and proadrenomedullin N-terminal 20 peptide on Hypertension 2003; 41: 512-8.
rat zona glomerulosa cells. Life Sci 1997; 60: 1693-7. [79] Wang X, Nishikimi T, Akimoto K, Tadokoro K, Mori Y, Minamino
[58] Mazzocchi G, Albertin G, Andrei PG, et al. Distribution, functional N. Upregulation of ligand, receptor system, and amidating activity of
role, and signaling mechanism of adrenomedullin receptors in the adrenomedullin in left ventricular hypertrophy of severely
rat adrenal gland. Peptides 1999; 20: 1479-87. hypertensive rats: effects of angiotensin-converting enzyme inhibitors
[59] Yamaguchi T, Baba K, Doi Y, Yano K. Effect of adrenomedullin and diuretic. J Hypertens 2003; 21: 1171-81.
on aldosterone secretion by dispersed rat adrenal zona glomerulosa [80] Dobrzynski E, Wang C, Chao J, Chao L. Adrenomedullin gene
cells. Life Sci 1995; 56: 379 - 87 delivery attenuates hypertension, cardiac remodeling, and renal
[60] Petrie MC, Hillier C, Morton JJ, McMurray JJ. Adrenomedullin injury in deoxycorticosterone acetate-salt hypertensive rats.
selectively inhibits angiotensin II-induced aldosterone secretion in Hypertension 2000; 36: 995-1001.
humans. J Hypertens 2000; 18: 61 - 64 [81] Nishikimi T, Yoshihara F, Horinaka S, et al. Chronic
[61] Yamaguchi T, Baba K, Doi Y, Yano K, Kitamura K, Eto, T. administration of adrenomedullin attenuates transition from left
Inhibition of aldosterone production by adrenomedullin, a ventricular hypertrophy to heart failure in rats. Hypertension 2003;
hypotensive peptide, in the rat. Hypertension 1996; 28: 308-14. 42: 1034-41.
[62] Samson WK, Murphy T, Schell DA. A novel vasoactive peptide, [82] Nakamura R, Kato J, Kitamura K, et al. Adrenomedullin
adrenomedullin, inhibits pituitary adrenocorticotropin release. administration immediately after myocardial infarction ameliorates
Endocrinology 1995; 136: 2349-52. progression of heart failure in rats. Circulation 2004; 110: 426-31.
Adrenomedullin in Heart Failure Current Hypertension Reviews, 2011, Vol. 7, No. 4 283

[83] Shimosawa T, Shibagaki Y, Ishibashi K, et al. Adrenomedullin, an [89] Rademaker MT, Charles CJ, Cooper GJ, et al. Combined
endogenous peptide, counteracts cardiovascular damage. Circulation endopeptidase inhibition and adrenomedullin in sheep with
2002; 105: 106-11. experimental heart failure. Hypertension 2002; 39: 93-8.
[84] Kawai J, Ando K, Tojo A, et al. Endogenous adrenomedullin [90] Nishikimi T, Mori Y, Ishimura K, et al. Chronic effect of combined
protects against vascular response to injury in mice. Circulation vasopeptidase inhibition and adrenomedullin in heart failure in rats.
2004; 109: 1147-53. Am J Hypertension 2006: 19: 1039-1048.
[85] Rademaker MT, Charles CJ, Lewis LK, et al. Beneficial [91] Nagaya N, Satoh T, Nishikimi T, et al. Hemodynamic, renal, and
hemodynamic and renal effects of adrenomedullin in an ovine hormonal effects of adrenomedullin infusion in patients with
model of heart failure. Circulation 1997; 96: 1983-1990. congestive heart failure. Circulation 2000;101: 498-503.
[86] Nagaya N, Nishikimi T, Horio T, et al. Cardiovascular and renal [92] Lainchbury JG, Nicholls MG, Espiner EA, Yandle TG, Lewis LK,
effects of adrenomedullin in rats with heart failure. Am J Physiol Richards AM. Bioactivity and interactions of adrenomedullin and
1999; 276: R213-R218. brain natriuretic peptide in patients with heart failure. Hypertension
[87] Rademaker MT, Charles CJ, Espiner EA, Nicholls MG, Richards 1999; 34: 70-5.
AM. Long-term adrenomedullin administration in experimental [93] Nishikimi T, Karasawa T, Inaba C, et al. Effects of Long-Term
heart failure. Hypertension 2002; 40: 667-72. Intravenous Administration of Adrenomedullin plus Human Atrial
[88] Rademaker MT, Charles CJ, Cooper GJ, et al. Combined Natriuretic Peptide Therapy in Acute Decompensated Heart
angiotensin-converting enzyme inhibition and adrenomedullin in an Failure: a pilot study. Circ J 2009; 73: 892-898.
ovine model of heart failure. Clin Sci 2002; 102: 653-60.

Received: July 01, 2011 Revised: August 30, 2011 Accepted: September 10, 2011

Das könnte Ihnen auch gefallen