You are on page 1of 7

articles

Angiotensin-converting enzyme 2 is an
essential regulator of heart function
Michael A. Crackower*†‡, Renu Sarao*§k, Gavin Y. Oudit‡k{#, Chana Yagilq, Ivona Kozieradzki*§, Sam E. Scanga**,
Antonio J. Oliveira-dos-Santos*, Joan da Costa*, Liyong Zhang*†‡§, York Pei#, James Scholey#, Carlos M. Ferrario††, Armen S. Manoukian**,
Mark C. Chappell††, Peter H. Backx‡{#, Yoram Yagilq & Josef M. Penninger*†‡§

* Amgen Research Institute/Ontario Cancer Institute; and † Departments of Medical Biophysics and Immunology; and ‡ The Heart & Stroke/Richard Lewar Centre for
Excellence in Cardiovascular Research; and { Department of Physiology; and # Department of Medicine and the University Health Network; and ** Division of Cellular
& Molecular Biology, University Health Network, Ontario Cancer Institute, University of Toronto, University Avenue, Toronto, Ontario M5G 2M9, Canada
§ IMBA, Institute for Molecular Biotechnology of the Austrian Academy of Sciences, c/o Dr Bohr Gasse, A-1030, Vienna, Austria
q Laboratory for Molecular Medicine, Department of Nephrology and Hypertension, Faculty of Health Science, Ben-Gurion University Barzilai Medical Center
Campus, Ashkelon 78306, Israel
†† The Hypertension and Vascular Disease Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1032, USA
k These authors contributed equally to the work
...........................................................................................................................................................................................................................

Cardiovascular diseases are predicted to be the most common cause of death worldwide by 2020. Here we show that angiotensin-
converting enzyme 2 (ace2) maps to a defined quantitative trait locus (QTL) on the X chromosome in three different rat models of
hypertension. In all hypertensive rat strains, ACE2 messenger RNA and protein expression were markedly reduced, suggesting that
ace2 is a candidate gene for this QTL. Targeted disruption of ACE2 in mice results in a severe cardiac contractility defect, increased
angiotensin II levels, and upregulation of hypoxia-induced genes in the heart. Genetic ablation of ACE on an ACE2 mutant
background completely rescues the cardiac phenotype. But disruption of ACER, a Drosophila ACE2 homologue, results in a severe
defect of heart morphogenesis. These genetic data for ACE2 show that it is an essential regulator of heart function in vivo.

Cardiovascular disease will be the greatest health care burden of the genetic quantitative trait loci (QTL) associated with hypertension in
twenty-first century1. A major risk factor for heart disease is high animal models4, none of these loci have been translated into genes5.
blood pressure2. Hypertension is a multifactorial quantitative trait Thus, the molecular and genetic mechanisms underlying hyperten-
controlled by both genetic and environmental factors3. While much sion and other cardiovascular diseases remain obscure.
is known about environmental factors that can contribute to high One important regulator of blood pressure homeostasis is the
blood pressure, such as diet and physical activity, less is known renin–angiotensin system (RAS). The protease renin cleaves angio-
about the genetic factors that are responsible for predisposition to tensinogen into the inactive decameric peptide angiotensin I
cardiovascular disease. Despite the identification of several putative (AngI). Angiotensin-converting enzyme (ACE) then catalyses the

Figure 1 Association of ACE2 and hypertension in the rat. a, Results of radiation hybrid Sabra SBH/y and SBN/y rats. c, Western blot analysis of ACE2 protein levels from kidneys
mapping of rat ace2, compared to the mapping of a QTL identified in Sabra salt-sensitive of Sabra SBH/y and their control SBN/y rats, as well as SHR and SHRSP and their control
animals (SS-X ), SHRSP (BP3 ), and SHR rats (BB.Xs). Polymorphic marker names are WKY rats. Systolic blood pressure in mm Hg for the respective Sabra rats is indicated. Bars
indicated to the left of the ideogram. LOD scores and theta values for markers linked to show mean values ^ s.e.m. Asterisk, P , 0.05; double asterisk, P , 0.01 (n ¼ 4, for
ace2 are shown. cR, centiradians. b, Northern blot analysis of ace2 mRNA from kidneys of all groups).

822 © 2002 Nature Publishing Group NATURE | VOL 417 | 20 JUNE 2002 | www.nature.com/nature
articles
cleavage of the AngI into the active octomer angiotensin II (AngII), the X chromosome with significant logarithm-of-odds (LOD)
which can contribute to hypertension by promoting vascular scores to markers DXRat9, DXWox14, DXWox15 and DXRat42,
smooth muscle vasoconstriction and renal tubule sodium reabsorp- placing ace2 between DXRat9 and DXRat42 (Fig. 1a). Comparative
tion6,7. ACE mutant mice display spontaneous hypotension, partial mapping showed that the ace2 map position overlaps with a QTL
male infertility, and kidney malformations8,9. In humans, an ACE interval for hypertension identified in Sabra salt-sensitive rats found
polymorphism has been associated with determinants of renal and between markers DXMgh12 and DXRat8 (SS-X) (ref. 18). Moreover,
cardiovascular function10, and pharmacological inhibition of ACE the chromosomal ace2 region maps to the BP3 QTL interval defined
and AngII receptors are effective in lowering blood pressure11 and in stroke-prone spontaneously hypertensive rats (SHRSP) rats19,
preventing kidney disease12. In addition, inhibition of ACE and and the hypertensive BB.Xs QTL identified on the X chromosome of
AngII receptors has beneficial effects in heart failure13. spontaneous hypertensive rats (SHR) by congenic analysis20 (Fig.
Recently, a homologue of ACE, termed ACE2, has been identified; 1a). Thus, ace2 maps to a QTL on the rat X chromosome that has
it is predominantly expressed in the vascular endothelial cells of the been identified in three separate models of spontaneous and diet-
kidney and heart14,15. Unlike ACE, ACE2 functions as a carboxy- induced hypertension.
peptidase, cleaving a single residue from AngI, generating Ang1-9
(refs 14, 15), and a single residue from AngII to generate Ang1-7
(ref. 14). These in vitro biochemical data suggest that ACE2 may Downregulation of ACE2 expression in hypertensive rats
modulate the RAS and thus affect blood pressure regulation. In Because the kidney is a major site of blood pressure regulation22, we
addition, it has been shown that ACE2 can cleave other peptide determined ace2 expression levels in the kidneys of these three
substrates14,15. Two different ACE2 homologues have been identified hypertensive rat strains. We initially measured ace2 mRNA levels in
in flies16,17. Nevertheless, the in vivo role of ACE2 in the cardiovas- the kidneys of salt-sensitive Sabra hypertensive (SBH/y) rats and
cular system and the RAS is not known. control salt-resistant Sabra normotensive (SBN/y) rats. Salt loading
(with deoxycorticosterone acetate (DOCA) salt) had no effect on
ACE2 maps to a QTL on the X chromosome in hypertensive rats ace2 mRNA expression in normotensive SBN/y rats. In SBH/y rats,
Hypertension and most cardiovascular diseases are multifactorial in salt loading and the development of hypertension were associated
nature and disease pathogenesis is influenced by multiple genetic with a significant reduction in ace2 mRNA expression compared to
susceptibility loci3. In various recombinant rat models, several QTL normotensive SBN/y rats (Fig. 1b). ace2 mRNA was also lower in
for hypertension have been identified. ace2 maps to the X chromo- SBH/y rats fed a regular diet (‘normal chow’) when compared to
some in human14 and a QTL has been mapped to the X chromo- SBN/y control rats fed a similar diet. This latter finding suggests that
some in several rat models of hypertension with no candidate gene downregulation of ACE2 may be independent of blood pressure.
ascribed to it as yet18–20. Angiotensin receptor II, which also maps to However, the possibility that ACE2 expression is controlled by
the X chromosome, has previously been excluded from this candi- blood pressure cannot be excluded.
date interval21. To measure ACE2 protein levels, we generated ACE2 (amino
We speculated that ace2 could be a candidate gene for this QTL. acids 206–225 of mouse ACE2) specific rabbit antiserum, which
Radiation hybrid mapping showed that the rat ace2 gene maps on cross-reacts with both rat and human ACE2 (not shown). In line
with the decreased ACE2 mRNA expression, ACE2 protein
expression was markedly reduced in SBH/y animals that were fed
a normal diet (Fig. 1c). Increase in blood pressure of SBH/y rats
following a 4-week diet of DOCA salt correlated with a further
decrease in ACE2 protein expression (Fig. 1c). Salt loading did not
trigger increased blood pressure23, nor did it alter ACE2 expression
in salt-resistant SBN/y control rats (Fig. 1c). ACE2 protein levels
were also significantly decreased in the kidneys of SHRSP and SHR
rats as compared to their WKY controls (Fig. 1c). Moreover, the
levels of ACE2 mRNA were markedly reduced in SHRSP and SHR
rats (not shown). Cloning and sequencing of the coding region of
ace2 in the hypertensive rat strains did not reveal any sequence
changes, indicating that reduced ACE2 expression probably results
from polymorphisms that control ace2 gene expression. The map
position and reduced expression of ACE2 in three different rat
strains indicate that ace2 is a strong candidate gene for this
hypertensive QTL on the X chromosome.

Generation of ACE2 knockout mice


To validate the candidacy of ace2 as a QTL and to test whether ACE2
Figure 2 ACE2-deficient mice. a, Western blot analysis of ACE2 protein expression in the has indeed an essential role in cardiovascular physiology and the
kidneys of ace2 þ/y ace 2/y mice. b, Polymerase chain reaction with reverse transcription pathogenesis of cardiovascular disease, we disrupted the ace2 gene
of RNA (RT–PCR) analysis of ACE mRNA expression in the heart and kidneys of ace2 þ/y in mouse by homologous recombination (see Supplementary
and ace2 2/y mice. c, Blood pressure measurements in 3-month-old ace2 þ/y (n ¼ 8) Information). The null mutation of ace2 was verified by the absence
and ace2 2/y (n ¼ 8) mice in the absence (left panels) or presence of the ACE blocker of ace2 mRNA transcripts and protein by northern (not shown) and
captopril. Mean values ^ s.e.m. are shown (double asterisk; P , 0.01). d, The top western blot analyses (Fig. 2a). ace mRNA expression in the kidneys
panels show haematoxylin and eosin stained sections of hearts isolated from 6-month-old and hearts was not altered in ace2 mutant mice (Fig. 2b). ACE2 null
ace2 þ/y and ace2 2/y mice. We note the enlarged left and right ventricles in ace2 2/y mice were born at the expected mendelian frequency, appeared
mice. However, the overall heart size is comparable between both genotypes and there is healthy, and did not display any gross detectable alterations in all
no evidence for cardiac hypertrophy macroscopically or in isolated cardiomyocytes). The organs analysed. Moreover, in contrast to ace 2/2 male mice that
bottom panels show an absence of interstitial fibrosis in ace2 2/y mice. As shown by PSR display significantly reduced fertility, both male and female ace2
staining (see Methods). null mice are fertile.

NATURE | VOL 417 | 20 JUNE 2002 | www.nature.com/nature © 2002 Nature Publishing Group 823
articles
Normal blood pressure in ace2 mutant mice Loss of ACE2 impairs heart function
It has been previously shown that ace mutant mice display reduced Pharmacological inhibition of ACE or AngII receptors suggested a
blood pressure and kidney pathology8,9. Therefore, we first tested role for the RAS in the regulation of heart function and cardiac
whether loss of ACE2 expression affects blood pressure homeostasis hypertrophy13. However, neither ace8,9 nor angiotensinogen24 null
and/or kidney development or function. Loss of ACE2 did not result mice develop any overt heart disease. ACE2 is highly expressed in
in alteration of blood pressure in 3-month-old ace2 2/y male (Fig. the vasculature of the heart, so we analysed hearts of ace2-deficient
2c) or ace2 2/2 female mice (not shown) as compared to their mice. Hearts of ace2 mutant mice display a slight wall thinning of
control littermates. Because it was possible that ACE could com- the left ventricle and increased chamber dimensions (Fig. 2d).
pensate for the loss of ACE2, we treated ace2-deficient mice with Thinning of the anterior left ventricular wall and increase in the
captopril, which blocks ACE but not ACE2 function14,15. However, left ventricle end diastolic dimension in ace2-deficient hearts can be
in vivo inhibition of ACE with captopril reduced the blood pressure also seen by echocardiography (Table 1). These structural changes
of ace 2/y male mice to a similar extent as was observed in captopril- are primarily observed in 6-month-old male mice. However, heart
treated wild-type littermates (Fig. 2c). Thus, even in a scenario of weights and heart to body weight ratios were comparable between
ACE inhibition, loss of ACE2 has no apparent direct effect on blood age-matched 3-month-old (not shown) and 6-month-old ace2 2/y
pressure homeostasis. In addition, no alterations in kidney ultra- and ace2 þ/y mice (see Supplementary Information). Echocardio-
structure or function or anaemia could be detected (not shown). graphy also showed that the left ventriclar-mass and its ratio to body
weight were normal (Table 1). We also failed to observe structural
and biochemical changes characteristic of dilated cardiomyopathy
as there was no indication of interstitial cardiac fibrosis (Fig. 2d) nor
prototypical changes in ANF, BNP, a-MHC, b-MHC and skeletal
muscle actin gene expression (not shown). In addition, individual
cardiomyocytes of ACE2 null mice exhibited no evidence of
hypertrophy and we did not observe any evidence of altered
cardiomyocyte apoptosis in ACE2 null mice as detected by TdT-
dependent dUTP-biotin nick end labelling (TUNEL) staining (not
shown). Thus, despite mild dilation of hearts in 6-month-old ACE2
null mice, there was no evidence of cardiac hypertrophy or dilated
cardiomyopathy.
Assessment of cardiac function by echocardiography revealed
that all ace2 2/y male and ace2 2/2 female mice exhibit severe
reduction in cardiac contractility as determined by decreased left
ventricle fractional shortening, and decreased velocity of circum-
ferential fibre shortening (Table 1 and Fig. 3a, b). The decrease in
function was found to be more severe in 6-month-old male than in
age-matched female mice. In addition, 3-month-old male mice had
a less pronounced phenotype than older animals (Table 1 and
Fig. 3b), suggesting a progression in the phenotype (Table 1).
Consistent with the decreased cardiac contractility, 6-month-old

Figure 3 Loss of ACE2 results in severe contractile dysfunction. a, Echocardiographic Figure 4 Upregulation of hypoxia markers and increased angiotensin II levels in the
measurements of contracting hearts in a 6-month-old ace2 þ/y mouse and two ace2 2/y absence of ACE2. a, b, Northern blot analysis of BNIP3 and PAI-1 mRNA expression
mice. Arrows indicate the distance between systolic contraction (LVESD) and diastolic levels, two hypoxia-inducible genes in 6-month-old ace2 þ/y and ace2 2/y male mice.
relaxation (LVEDD). b, Per cent fractional shortening and velocity of circumferential fibre a, Individual northern blot data; b, relative levels of BNIP3 and PAI-1 mRNA levels
shortening (circumferences s21) in 6-month-old ace2 þ/y (n ¼ 8) and ace2 2/y (n ¼ 8) normalized to the gapdh control. Double asterisk, P , 0.01 (n ¼ 5). c, Angiotensin I
mice and 6-month-old ace2þ/2 (n ¼ 5) and ace2 2/2 (n ¼ 5) female mice. c, Tail-cuff (AngI), and Angiotensin II (AngII) peptide levels in the heart, kidneys and plasma of male
blood pressure measurements in 6-month-old male ace2 þ/y (n ¼ 8) and ace2 2/y ace2 þ/y (n ¼ 8) and ace2 2/y (n ¼ 8) littermate mice. Peptide levels were determined by
(n ¼ 8) mice and 6-month-old female ace2 þ/2 (n ¼ 5) and ace2 2/2 (n ¼ 5) mice. radioimmunoassays. Mean peptide levels ^ s.e.m. are shown. Double asterisk,
Mean values ^ s.e.m. are shown. Asterisk, P , 0.05 and double asterisk, P , 0.01. P , 0.01.

824 © 2002 Nature Publishing Group NATURE | VOL 417 | 20 JUNE 2002 | www.nature.com/nature
articles
male ace2 2/y mice exhibited reduced blood pressure (Fig. 3c), a
feature not found in age-matched ace2 2/2 females and 3-month-
old males. This suggests that the reduction in blood pressure may be
the result of severe cardiac dysfunction and not a direct effect of loss
of ACE2 on systemic blood pressure. To confirm the echocardi-
graphic defects in cardiac function, invasive haemodynamic
measurements were performed in ace2 null mice. These invasive
haemodynamic measurements showed that both the maximum and
minimum rates of change of left ventricular pressure, (dP/dt)max
and (dP/dt)min, were markedly reduced in the ace2 mutant mice
(Table 1), indicating severe impairment of contractile heart func-
tion. Loss of ACE2 also resulted in a significant decrease in aortic
and ventricular pressures consistent with the observed reductions in
cardiac contractility (data not shown). These results show that
ACE2 is an important regulator of heart function in vivo.

Upregulation of hypoxia-inducible genes in ace2 null mice


The severe contractile dysfunction and mild dilation in the absence
of hypertrophy or cardiac fibrosis in ace2 null mice resembles
cardiac stunning/hibernation in human and animal models25,26.
Cardiac stunning and hibernation are adaptive responses to chronic
hypoxia such as in coronary artery disease or following bypass
surgery27. Because ACE2 is highly expressed in vascular endothelial
cells but contractility is controlled by cardiomyocytes, we speculated
that loss of ACE2 could result in cardiac hypoxia. We therefore
analysed changes in the expression levels of hypoxia-inducible genes
such as BNIP3 (ref. 28) and PAI-1 (ref. 29) by northern blotting. In
the hearts of all ace2 null mice analysed, mRNA expression of BNIP3
and PAI-1 was markedly upregulated compared to their wild-type
littermates (Fig. 4a and b). The magnitude of increased expression
of these markers of hypoxia is similar to that previously observed in
other hypoxic models such as in myocyte-specific vascular endo-
thelial growth factor mutant mice30. Thus, loss of ACE2 results in
the induction of a hypoxia-regulated gene expression profile.

Increased angiotensin II levels in ace2 null mice


Because ACE2 functions as a carboxypeptidase, cleaving a single
residue from AngI to generate Ang1-9 (refs 14, 15) and a single
residue from AngII to generate Ang1-7 (ref. 14), we hypothesized Figure 5 ACE/ACE2 double mutant mice do not develop cardiac dysfunction. a, Blood
that ACE2 may regulate the RAS by competing with ACE for the pressure measurements in 6-month-old ace2 þ/y (n ¼ 8), ace2 2/y (n ¼ 8), ace 2/2
substrate AngI and/or cleaving and inactivating AngII. If correct, (n ¼ 8), and ace 2/2/ace2 2/y double mutant (n ¼ 6) male mice. b, Per cent fractional
loss of ACE2 should increase AngII levels in vivo. Using radio- shortening and velocity of circumferential fibre shortening in 6-month-old male ace2 þ/y
immunoassays, AngII levels were indeed found to be significantly (n ¼ 8), ace2 2/y (n ¼ 8), ace 2/2 (n ¼ 8) and ace 2/2/ace2 2/y double mutant (n ¼ 6)
increased in the kidneys, hearts and plasma of ace2 mutant mice littermates. c, Echocardiographic measurements of contracting hearts in 6-month-old
(Fig. 4c). In addition, an increase in AngI was observed in the heart male ace2 þ/y, ace2 2/y, ace 2/2 and ace 2/2/ace2 2/y double mutant littermate mice.
and kidney of ace2 mutant mice (Fig. 4c), consistent with AngI Mean values ^ s.e.m. are shown. Asterisk, P , 0.05 and double asterisk, P , 0.01.

Table 1 Heart functions of ace2 null mice


3-month-old males 6-month-old males 6-month-old females
ace2 þ/y ace2 2/y ace2 þ/y ace2 2/y ace2 þ/2 ace2 2/2
...................................................................................................................................................................................................................................................................................................................................................................
n¼7 n¼7 n¼8 n¼8 n¼3 n¼3
Heart rate (b.p.m.) 469 ^ 12 466 ^ 18 495 ^ 15 482 ^ 12 460 ^ 6 452 ^ 14
Anterior left ventricular wall thickness (mm) 0.65 ^ 0.02 0.62 ^ 0.01 0.66 ^ 0.01 0.59 ^ 0.02* 0.65 ^ 0.02 0.57 ^ 0.04
LVEDD (mm) 4.09 ^ 0.04 4.25 ^ 0.10 4.12 ^ 0.10 4.49 ^ 0.12* 3.71 ^ 0.10 1.11 ^ 0.07
LVESD (mm) 2.11 ^ 0.04 2.69 ^ 0.09** 2.13 ^ 0.06 3.29 ^ 0.14** 1.74 ^ 0.07 2.68 ^ 0.11**
Fractional shortening (%) 48.42 ^ 1.14 36.73 ^ 0.89** 48.12 ^ 0.84 26.76 ^ 1.78** 53.00 ^ 1.68 34.85 ^ 1.76**
Peak aortic outflow velocity (m s21) 0.992 ^ 0.029 0.922 ^ 0.033 0.902 ^ 0.044 0.802 ^ 0.037* 0.875 ^ 0.048 0.809 ^ 0.044
Velocity of circumferential fibre shortening (circ s21) 9.49 ^ 0.48 7.28 ^ 0.45* 9.46 ^ 0.26 4.93 ^ 0.33** 10.34 ^ 0.81 6.16 ^ 0.31**
Calculated left ventricular mass (mg) 95.69 ^ 1.88 95.50 ^ 2.28 102.41 ^ 4.00 100.49 ^ 3.39 92.27 ^ 2.02 89.60 ^ 1.54
Left ventricular mass/body mass (mg g21) 3.32 ^ 0.08 3.53 ^ 0.18 3.19 ^ 0.10 3.32 ^ 0.15 3.29 ^ 0.37 3.30 ^ 0.09

(dP/dt)max n.d. n.d. 5,579 ^ 422 3,034 ^ 124** n.d. n.d.


(dP/dt)min n.d. n.d. 25,055 ^ 257 22,929 ^ 271** n.d. n.d.
...................................................................................................................................................................................................................................................................................................................................................................
*P , 0.05 ace2 2/y versus ace2 þ/y or ace2 2/2 versus ace2 þ/2.
**P , 0.01 ace2 2/y versus ace2 þ/y or ace2 2/2 versus ace2 þ/2.
b.p.m., heart beats per minute; LVEDD, left ventricle end diastolic dimension; LVESD, left ventricle end systolic dimension; (dP/dt)max, maximum first derivative of the change in left ventricular pressure
with time; (dP/dt)min, minimum first derivative of the change in left ventricular pressure with time. n.d. not determined.

NATURE | VOL 417 | 20 JUNE 2002 | www.nature.com/nature © 2002 Nature Publishing Group 825
articles
being a substrate of ACE2 action in vivo. No differences in ACE Supplementary Information). Both fly homologues are expressed in
mRNA levels were found in the hearts and kidneys of ace2 mutant developing heart cells of the Drosophila embryo16,17. To determine
mice compared to controls, indicating that the increased AngII what role these ACE homologues may have in heart development,
tissue levels were not due to increased ACE expression (Fig. 2b). we studied mutant flies that carry a P-element insertion in the Acer
These data show that ACE2 functions as a regulator of the RAS locus (P679) (ref. 31). Mutation of ACER results in early embryonic
modulating endogenous levels of AngI and AngII. lethality (not shown). We, therefore, examined heart tube morpho-
genesis using two early markers (the even-skipped (Eve) and Tin-
Ablation of ACE expression rescues ace2-deficient mice man (Tin) proteins) for heart progenitor cells in Drosophila
To determine whether genetic ablation of ACE in combination with embryos.
disruption of ACE2 has any effect on the heart phenotype in ace2 Expression of Eve is the earliest available marker for heart
mutant mice, we generated ace/ace2 double mutant mice. These progenitor cells32,33 and in wild-type stage-11 embryos, clusters of
double mutant mice were born at the expected mendelian ratio and three Eve-positive heart progenitor cells form the earliest heart tube
appear healthy. Blood pressure (Fig. 5a) and kidney defects (not anlage (Fig. 6a). By contrast, ACER mutant flies display reduced
shown) in the ace/ace2 double null mice were similar to that of ace numbers and disorganization of Eve positive progenitor cells (Fig.
single mutant mice. Fertility of the ace/ace2 double mutant mice was 6b). To study further whether ACER mutant mice have a defect in
not addressed. Thus, loss of both ACE and ACE2 does not cause any mesoderm that specifies heart progenitor cells, we used Tinman as a
apparent disease in addition to that seen in ace single mutant mice. marker. Tinman provides the dorsal mesoderm with the compe-
The heart function of ACE knockout mice has not been pre- tence to specify heart progenitors34, a role conserved in tinman-
viously reported, to our knowledge, so we first analysed the heart related homeobox genes such as murine Nkx2.5 that is essential for
parameters in these mice. In ace 2/2 mice, hearts are histologically heart development35. Similar to alterations in Eve expression, loss of
normal (not shown) and no defect in heart function could be ACER results in reduced numbers and disorganization of the Tin-
detected at 6 months of age (Fig. 5b, c). However, ablation of ACE man positive dorsal mesoderm (Fig. 6c, d). Thus, ACER mutant flies
expression on an ace2 mutant background completely abolished the have defective heart morphogenesis and ACER may have a role in
cardiac dysfunction phenotype of ace2 single knockout mice the specification of heart progenitors in Drosophila embryos.
(Fig. 5b, c). Using echocardiography, all heart functions of
6-month-old, age-matched ace/ace2 double mutant mice were Discussion
comparable to that of their ace single mutant and wild-type Here we have shown that ace2 maps to a QTL associated with
littermates (Fig. 5, Table 1 and Supplementary Information). hypertension in three rat models of high blood pressure. ACE2 levels
Restoration of heart functions occurred in both male and female are reduced in all of these hypertensive rat strains. In mice, genetic
ace/ace2 double mutant mice. Importantly, there was also no inactivation of ACE2 using homologous recombination results in
difference in blood pressure between ace and ace/ace2 knockout increased AngII peptide levels, upregulation of hypoxia genes in the
mice (Fig. 5a), further implying that the reduced blood pressure in heart and severe cardiac dysfunction. Ablation of ACE expression
older male ace2 mice is due to the dramatic decrease in heart on an ace2-deficient background completely abolished the heart-
function. failure phenotype of ace2 single knockout mice. Our results in
Drosophila show that disruption of the fly ACE2 homologue,
Heart defects in ACER mutant flies ACER17, results in a severe and lethal defect of heart morphogenesis.
ACE2 and ACE have critical roles in heart function in mice, so we Our fly data also indicate that ACER has a role in the specification of
analysed whether the role of ACE2 in the heart is evolutionarily heart progenitor cells. Thus, fly ACER and mammalian ACE/ACE2
conserved. Flies have two ACE2 homologues called ANCE16 and may have divergent roles in heart progenitor cell specification versus
ACER17, both of which have a domain structure similar to ACE2 (see the control of heart function.
Most cardiovascular diseases are multifactorial quantitative traits
controlled by both genetic and environmental factors3. One major
factor for cardiovascular disease is the RAS. The map position and
reduced expression of ACE2 in three different rat strains indicate
that ace2 is a strong candidate gene for a hypertensive QTL on the X
chromosome. However, loss of ACE2 in mice has no apparent effect
on blood pressure even when ACE is inhibited. From previous
studies in many systems, it has become clear that the interplay of
several QTLs determine the phenotype. Currently it is thought that
there are numerous genetic polymorphisms that serve to regulate
blood pressure in humans and rodents3. Thus, altering one of the
genes in the ‘wrong’ genetic mouse background would not be
sufficient in itself to alter blood pressure. Only when a sufficient
number of control mechanisms are altered in concert would high
blood pressure ensue. But despite the lack of blood pressure changes
in the ACE2 mutant mice, there is an increase in AngII levels. These
data clearly indicate that ACE2 controls the levels of AngII in vivo,
supporting the candidacy of ace2 as a QTL that controls the RAS.
Figure 6 Expression of heart progenitor markers in Drosophila ACER mutant embryos. Thus, the absence of increases in blood pressure in ACE2 mutant
a, b, Expression of the heart progenitor cell marker even-skipped (Eve) in stage-11 wild- mice does not exclude ace2 as a candidate gene for the QTL on the X
type and ACER mutant fly embryos. Bottom panels show close-ups of the Eve-positive chromosome. It will be interesting to determine whether single
heart progenitor cells. Arrows in a show clusters of three Eve-positive heart progenitor nucleotide polymorphisms in the human ACE2 locus correlate with
cells in wild-type embryos. In ACER mutant embryos, there is disorganization and changes in blood pressure.
reduction of Eve-positive cells (arrows). c, d, Expression of Tinman (Tin), a specification Unexpectedly, loss of ACE2 in mice results in profound contrac-
marker for heart progenitor cells, in stage-12 embroys. In ACER mutant embryos, the tile dysfunction. The complete rescue of the heart phenotype in
numbers of Tin-expressing cells is markedly reduced in the dorsal mesoderm as ACE/ACE2 double mutant mice indicates that ACE expression has a
compared to wild-type Drosophila embryos. causative role in the onset of the heart dysfunction. Both ACE and

826 © 2002 Nature Publishing Group NATURE | VOL 417 | 20 JUNE 2002 | www.nature.com/nature
articles
ACE2 have been shown to be able to cleave AngI in vitro6,7,14,15. (1 mm Hg ¼ 133.3 Pa) and subsequently embedded in paraffin. Myocardial interstitial
fibrosis was determined by quantitative morphometry using the colour-subtractive
Whereas ACE generates AngII from AngI, ACE2 can counteract the computer-assisted image analysis using Image Processing Tool Kit version 2.5 coupled
function of ACE by cleaving AngII or by competing with ACE for with Photoshop 6.0 software. Picro-Sirius-red (PSR)-stained sections were used to
the substrate AngI. Thus, our mouse data suggest that increased calculate interstitial fibrosis as the ratio of the areas with positive PSR staining compared
AngII mediates the heart phenotype. Nevertheless, our data does to the entire visual field. Echocardiographic assessments were performed as described39.
not exclude the possibility that other metabolites6,7,14,15 of ACE2 and Mice were anaesthetized with isoflurane/oxygen (1.25%/98.75%) and examined by
transthoracic echocardiography using a Acuson Sequoia C256 equipped with a 15-MHz
ACE contribute to the observed phenotypes. It will be important to linear transducer. Haemodynamics measurements were performed as described40. Tail-
assess directly the role of AngII in the heart defects of ACE2 mutant cuff blood pressure measurement were taken using a Visitech BP-2000 Blood Pressure
mice by pharmacological or genetic inhibition of AngII receptors, Analysis System manufactured by Visitech Systems (Apex). For captopril treatment,
and determine if any other ACE2-modulating peptides are altered in drinking water was supplemented with 400 mg l21 captopril (Sigma) for two weeks before
blood pressure measurement.
these mice. We note that in human patients, inhibition of ACE or
AngII receptors can improve the outcome of heart failure13. The Plasma and tissue angiotensins
contribution of ACE2 in human heart failure needs to be For plasma measurements blood was collected into chilled Vacutainer blood collection
determined. tubes (Becton Dickinson) containing a mixture of peptidase inhibitors (25 mM
The heart phenotype found in ACE2 mutant mice is similar to ethylenediaminetetraacetic acid (EDTA), 0.44 mM o-phenanthroline (PHEN), 1 mM 4-
chloromercuribenzoic acid (PCMB), 0.12 mM pepstatin A and 3 mM acetyl-His-Pro-Phe-
that seen in cardiac stunning/hibernation such as in humans with Val-Statine-Leu-Phe, a renin inhibitor. Frozen hearts and kidneys (stored at 280 8C) were
coronary artery disease and cases of bypass surgery25–27. In humans homogenized on ice in 80% ethanol/0.1 M HCl containing the peptidase inhibitors
and in animal models of cardiac stunning/hibernation, chronic described above including phenylmethylsulphonyl fluoride (PMSF, 100 mM). A sample of
hypoxic conditions lead to compensatory changes in myocyte the homogenate was taken to determine total protein content, using the Bradford protein
assay with bovine serum albumin as a standard (BioRad Protein Assay Reagent, BioRad
metabolism27, upregulation of hypoxia-induced genes30, and severe Laboratories). Peptide extraction was performed as previously described41.
contractile dysfunction25–27,30. Because ACE2 is expressed in the Radioimmunoassay analysis of angiotensin peptide content in the extracts from plasma,
vascular endothelium and not in cardiac myocytes15, local increases heart and kidney tissue was performed as previously described42. The limits of detection
in AngII may lead to vasoconstriction, resulting in hypoperfusion for the AngII, and AngI radio immunoassays were 0.5 fmol per tube, and 5 fmol per tube,
respectively.
and hypoxia in the myocardium. It has been established that AngII
can induce oxidative stress in endothelial cells36,37, so the increase in Drosophila studies
AngII could also result in dysfunction of the vascular endothelium The P-element allele of ACER, P679 (ref. 31), was used to generate homozygous ACER
of the heart via the induction of oxidative stress. A mutant embryos. Whole embryo staining with Eve and Tin antibodies was as described33.
Received 6 March; accepted 8 April 2002; doi:10.1038/nature00786.
Methods 1. Yusuf, S., Reddy, S., Ounpuu, S. & Anand, S. Global burden of cardiovascular diseases. Part I: General
Cloning of mouse and rat ACE2 and chromosomal QTL mapping considerations, the epidemiologic transition, risk factors, and impact of urbanization. Circulation
Murine ace2 was cloned from a proprietary EST database. Using a mouse ace2 probe, we 104, 2746–2753 (2001).
then screened a rat kidney cDNA (Invitrogen) to obtain a full-length rat cDNA as 2. Carretero, O. A. & Oparil, S. Essential hypertension. Part I: Definition and etiology. Circulation 101,
determined by DNA sequencing (see Supplementary Information). For chromosomal 329–335 (2000).
mapping, a rat ace2 cDNA specific probe was used to screen a rat PAC library (RPCI-31, 3. Jacob, H. J. Physiological genetics: Application to hypertension research. Clin. Exp. Pharm. Phys. 26,
Research Genetics), identifying two positive clones (6M6 and 125K9). The end sequences 530–535 (1999).
4. Rapp, J. P. Genetic analysis of inherited hypertension in the rat. Physiol. Rev. 80, 135–172 (2000).
of these clones were determined and rat-specific primers were designed (mc2L: 5 0 -
5. Stoll, M. et al. A genomic-systems biology map for cardiovascular function. Science 294, 1723–1726
TCAATTTACTGCTGAGGGGG-3 0 ; and mc2R: 5 0 -GAGGGATAACCCAGTGCAAA-3 0 )
(2001).
to determine the chromosomal map position of ACE2 in rat by screening a radiation
6. Corvol, P. & Williams, T. A. in Handbook of Proteolytic Enzymes (eds Barrett, A. J., Rawlings, N. D. &
hybrid panel (RH07.5, Research Genetics). SHR and control Wistar Kyoto (WKY) rats
Woessner, J. F.) 1066–1076 (Academic, London, 1998).
were obtained from Harlan and maintained at the animal facilities of the Ontario Cancer
7. Skeggs, L. T., Dorer, F. E., Levine, M., Lentz, K. E. & Kahn, J. R. The biochemistry of the renin-
Institute in accordance with institutional guidelines. Salt-resistant and salt-sensitive Sabra
angiotensin system. Adv. Exp. Med. Biol. 130, 1–27 (1980).
rats were bred and maintained at the animal facility of the Ben-Gurion University, Barzilai
8. Krege, J. H. et al. Male–female differences in fertility and blood pressure in ACE-deficient mice. Nature
Medical Center, Israel. DOCA salt treatment was as described previously18. 375, 146–148 (1995).
9. Esther, C. R. et al. Mice lacking angiotensin-converting enzyme have low blood pressure, renal
Expression analysis pathology and reduced male fertility. Lab. Invest. 74, 953–965 (1996).
Total RNAwas prepared from rat kidneys using tri-reagent. 20 mg of RNAwas resolved on a 10. Wuyts, B., Delanghe, J. & De Buyzere, M. Angiotensin I-converting enzyme insertion/deletion
0.8% formamide gel, blotted to nylon membrane (Amersham) and probed with a partial polymorphism: clinical implications. Acta Clin. Belg. 52, 338–349 (1997).
rat ACE2 cDNA clone (9-1). The b-actin probe and multiple-tissue northern blots were 11. Elkind, M. S. & Sacco, R. L. Stroke risk factors and stroke prevention. Semin. Neurol. 18, 429–440
purchased from Clontech. For western analysis, kidneys were homogenized in lysis buffer (1998).
(50 mM Tris-HCl, pH 7.4, 20 mM EDTA, and 1% triton-X100) supplemented with 12. Hollenberg, N. K. Angiotensin converting enzyme inhibition and the kidney. Curr. Opin. Cardiol. 3
“Complete” protease inhibitor cocktail (Roche) and 1 mM Na3VO4. 100 mg of protein was (Suppl. 1), S19–S29 (1988).
resolved by SDS–polyacrylamide gel electrophoresis (PAGE) on 8% tris-glycine gels. ACE2 13. Garg, R. & Yusuf, S. Overview of randomized trials of angiotensin-converting enzyme inhibitors on
immuno-serum was obtained from rabbits immunized with a mouse-specific ACE2 mortality and morbidity in patients with heart failure. J. Am. Med. Assoc. 273, 1450–1456 (1995).
peptide: DYEAEGADGYNYNRNQLIED. The serum was affinity-purified with the 14. Tipnis, S. R. et al. A human homolog of angiotensin-converting enzyme. Cloning and functional
immunizing peptide using sulpho-link kit (Pierce). A commercially available b-actin expression as a captopril-insensitive carboxypeptidase. J. Biol. Chem. 275, 33238–33243 (2000).
antibody was used as loading control (Santa Cruz). 15. Donoghue, M. et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2)
converts angiotensin I to angiotensin 1-9. Circ. Res. 87, e1–e8 (2000).
Generation of ACE2 mutant mice 16. Cornell, M. J. et al. Cloning and expression of an evolutionary conserved single-domain angiotensin
converting enzyme from Drosophila melanogaster. J. Biol. Chem. 270, 13613–13619 (1995).
A targeting vector (559 base pair short arm and 8.1 kilobase long arm) was constructed
17. Taylor, C. A., Coates, D. & Shirras, A. D. The Acer gene of Drosophila codes for an angiotensin-
using the pKO Scrambler NTKV-1907 vector (Stratagene). A portion of the ace2 genomic
converting enzyme homologue. Gene 181, 191–197 (1996).
DNA containing nucleotides þ1069 to þ1299 was replaced with the neomycin resistance
18. Yagil, C. et al. Role of chromosome X in the Sabra rat model of salt-sensitive hypertension.
cassette in the anti-sense orientation. The targeting construct was electroporated into
Hypertension 33 Part II, 261–265 (1999).
E14K embryonic stem cells, and screening for positive homologous recombinant
19. Hilbert, P. et al. Chromosomal mapping of two genetic loci associated with blood-pressure regulation
embryonic stem cell clones was performed by Southern blotting of EcoRI-digested
in hereditary hypertensive rats. Nature 353, 521–529 (1991).
genomic DNA hybridized to 5 0 and 3 0 flanking probes. Two independent ace 2/y
20. Kloting, I., Voigt, B. & Kovacs, P. Metabolic features of newly established congenic diabetes-prone
embryonic stem cell lines were injected into C57BL/6-derived blastocysts to generate BB.SHR rat strains. Life Sci. 62, 973–979 (1998).
chimaeric mice, which were backcrossed to C57BL/6 mice. Two embryonic stem cell lines 21. Koike, G. et al. Cloning, characterization, and genetic mapping of the rat type 2 angiotensin II receptor
gave independent germline transmission. Histology of all tissues, apoptopsis assays, blood gene. Hypertension 26, 998–1002 (1995).
serology and kidney morphometries were as described38. Complete ACE mutant mice have 22. Laragh, J. H. Renovascular hypertension: a paradigm for all hypertension. J. Hypertens. 4 (Suppl. 4),
been previously described8 and were obtained from Jackson Laboratories. Mice were S79–S88 (1986).
maintained at the animal facilities of the Ontario Cancer Institute in accordance with 23. Yagil, C. et al. Development, genotype and phenotype of a new colony of the Sabra hypertension prone
institutional guidelines. (SBH/y) and resistant (SBN/y) rat model of salt sensitivity and resistance. J. Hypertens. 14, 175–182
(1996).
Heart parameter measurements 24. Tanimoto, K. et al. Angiotensinogen-deficient mice with hypotension. J. Biol. Chem. 269,
For heart morphometry, hearts were perfused with 10% buffered formalin at 60 mm Hg 31334–31337 (1994).

NATURE | VOL 417 | 20 JUNE 2002 | www.nature.com/nature © 2002 Nature Publishing Group 827
articles
25. Kloner, R. A., Bolli, R., Marban, E., Reinlib, L. & Braunwald, E. Medical and cellular implications of 39. Wickenden, A. D. et al. Targeted expression of a dominant-negative K(v)4.2 K(þ) channel subunit in
stunning, hibernation, and preconditioning: and NHLBI workshop. Circulation 97, 1848–1867 the mouse heart. Circ. Res. 85, 1067–1076 (1999).
(1998). 40. Zvaritch, E. et al. The transgenic expression of highly inhibitory monomeric forms of phospholamban
26. Murphy, A. M. et al. Transgenic mouse model of stunned myocardium. Science 287, 488–491 (2000). in mouse heart impairs cardiac contractility. J. Biol. Chem. 275, 14985–14991 (2000).
27. Heusch, G. Hibernating myocardium. Physiol. Rev. 78, 1055–1085 (1998). 41. Allred, A. J., Chappell, M. C., Ferrario, C. M. & Diz, D. I. Differential actions of renal ischemic injury
28. Sowter, H. M., Ratcliffe, P. J., Watson, P., Greenberg, A. H. & Harris, A. L. HIF-1-dependent regulation on the intrarenal angiotensin system. Am. J. Physiol. Renal 279, F636–F645 (2000).
of hypoxic induction of the cell death factors BNIP3 and NIX in human tumors. Cancer Res. 61, 42. Chappell, M. C., Milsted, A., Diz, D. I., Brosnihan, K. B. & Ferrario, C. M. Evidence for an intrinsic
6669–6673 (2001). angiotensin system in the canine pancreas. J. Hypertens. 9, 751–759 (1991).
29. Kietzmann, T., Roth, U. & Jungermann, K. Induction of the plasminogen activator inhibitor-1 gene
expression by mild hypoxia via a hypoxia response element binding the hypoxia-inducible factor-1 in Supplementary Information accompanies the paper on Nature’s website
rat hepatocytes. Blood 94, 4177–4185 (1999). (http://www.nature.com/nature).
30. Giordano, F. J. et al. A cardiac myocyte vascular endothelial growth factor paracrine pathway is
required to maintain cardiac function. Proc. Natl Acad. Sci. USA 98, 5780–5785 (2001).
31. Spradling, A. C. et al. The Berkeley Drosophila Genome Project gene disruption project: Single P- Acknowledgements
element insertions mutating 25% of vital Drosophila genes. Genetics 153, 135–177 (1999). We thank D. Ganten for supplying us with tissue from SHRSP rats. Eve and Tin antibodies
32. Frasch, M., Hoey, T., Rushlow, C., Doyle, H. J. & Levine, M. Characterization and localization of the were a gift from M. Frasch. We acknowledge the Samuel Lunenfeld Research Institute’s
even-skipped protein of Drosophila. EMBO J. 6, 749–759 (1987). CMHD Mouse Physiology Facility for their technical screening services. This study was
33. Azpiazu, N., Lawrence, P., Vincent, J-P. & Frasch, M. Segmentation and specification of the Drosophila supported by Amgen and by grants from the Israel Science Foundation and the German–
mesoderm. Genes Dev. 10, 3183–3194 (1996). Israeli Foundation for Scientific Research and Development to C.Y. and Y.Y. J.M.P. holds a
34. Zhizhang, Y. & Frasch, M. Regulation and function of tinman during dorsal mesoderm induction and Canadian Research Chair in Cell Biology. M.A.C. is supported in part by a Canadian
heart specification in Drosophila. Dev. Gen. 22, 187–200 (1998). Institutes of Health Research fellowship.
35. Harvey, R. NK-2 homeobox genes and heart development. Dev. Biol. 178, 203–216 (1996).
36. Cai, H. & Harrison, D. G. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress.
Circ. Res. 87, 840–844 (2000).
37. Enseleit, F., Hurlimann, D. & Luscher, T. F. Vascular protective effects of angiotensin converting Competing interests statement
enzymes inhibitors and their relation to clinical events. J. Cardiovasc. Pharmacol. 37 (Suppl. 1), The authors declare that they have no competing financial interests.
S21–S30 (2001).
38. Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph- Correspondence and requests for materials should be addressed to J.M.P.
node organogenesis. Nature 397, 315–323 (1999). (e-mail: jpenning@uhnres.utoronto.ca).

828 © 2002 Nature Publishing Group NATURE | VOL 417 | 20 JUNE 2002 | www.nature.com/nature

You might also like