• No results found

The renin angiotensin system (RAS), a potent regulator of

N/A
N/A
Protected

Academic year: 2021

Share "The renin angiotensin system (RAS), a potent regulator of"

Copied!
7
0
0

Loading.... (view fulltext now)

Full text

(1)

Angiotensin-(1-7) Receptor Mas Knockout Mice

Robson A.S. Santos, Carlos H. Castro, Elisandra Gava, Se´rgio V.B. Pinheiro, Alvair P. Almeida,

Renata Dutra de Paula, Jader S. Cruz, Anderson S. Ramos, Kaleizu T. Rosa, Maria C. Irigoyen,

Michael Bader, Natalia Alenina, Gregory T. Kitten, Anderson J. Ferreira

Abstract—In this study we investigated the effects of the genetic deletion of the angiotensin (Ang)-(1-7) receptor Mas on heart function. Localization of Mas in the mouse heart was evaluated by binding of rhodamine-labeled Ang-(1-7). Cardiac function was examined using isolated heart preparations. Echocardiography was used to confirm the results obtained with isolated heart studies. To elucidate the possible mechanisms involved in the cardiac phenotype observed in Mas⫺/⫺mice, whole-cell calcium currents in cardiomyocytes and the expression of collagen types I, III, and VI and

fibronectin were analyzed. Ang-(1-7) binding showed that Mas is localized in cardiomyocytes of the mouse heart. Isolated heart techniques revealed that Mas-deficient mice present a lower systolic tension (average: 1.4⫾0.09 versus 2.1⫾0.03 g in Mas⫹/⫹mice),dT/dt, and heart rate. A significantly higher coronary vessel resistance was also observed

in Mas-deficient mice. Echocardiography revealed that hearts of Mas-deficient mice showed a significantly decreased fractional shortening, posterior wall thickness in systole and left ventricle end-diastolic dimension, and a higher left ventricle end-systolic dimension. A markedly lower global ventricular function, as defined by a higher myocardial performance index, was observed. A higher delayed time to the peak of calcium current was also observed. The changes in cardiac function could be partially explained by a marked change in collagen expression to a profibrotic profile in Mas-deficient mice. These results indicate that Ang-(1-7)-Mas axis plays a key role in the maintenance of the structure and function of the heart.(Hypertension. 2006;47:996-1002.)

Key Words: receptors, angiotensin 䡲 extracellular matrix 䡲 echocardiography 䡲 heart failure 䡲 cardiac function

T

he renin–angiotensin system (RAS), a potent regulator of blood pressure (BP), plays a major role in the pathogen-esis of cardiovascular diseases.1–3 Although angiotensin

(Ang) II is the major effector of this system, several other Angs are now recognized as being biologically active. The heptapeptide Ang-(1-7) is particularly interesting, because it appears to counterbalance most of the Ang II effects. Thus, Ang-(1-7) may have a pivotal role in the cardiovascular sys-tem.4 –7 Accordingly, Ang-(1-7) has been reported to present

important effects in coronary vessels and heart function.8 –14This

peptide produced a significant increase in cardiac output and stroke volume in anesthetized Wistar rats,15 decreased the

incidence and duration of ischemia-reperfusion arrhythmias,11

apparently by activating the sodium pump,14and improved the

postischemic contractile function12 in isolated perfused rat

hearts. In keeping with these data, chronic infusion of Ang-(1-7) improved endothelial aortic function and coronary perfusion and preserved cardiac function in a rat model of heart failure.10More

recently, using a transgenic rat that expresses an

Ang-(1-7)-producing fusion protein, an improvement in cardiac function was observed.16Ang-(1-7) also reduces the overall growth of

cardiomyocytes.16 –18 Concerning the effects in the coronary

vasculature, it has been shown that Ang-(1-7) elicits vasorelax-ation in isolated canine8and porcine19coronary artery rings. In

isolated perfused mouse hearts, Ang-(1-7) produced complex vascular effects involving interaction of its receptor with angio-tension II type 1 (AT1) and AT2receptor–related mechanisms,

leading to vasodilation.20

We have reported recently that Ang-(1-7) is an endogenous ligand for the G protein-coupled receptor Mas.21The genetic

deletion ofMasabolished the binding of Ang-(1-7) to mouse kidneys and abrogated the antidiuretic effect of Ang-(1-7) in mice after acute water load. In addition, aortic rings from Mas-deficient mice lack the endothelium-dependent Ang-(1-7)-induced relaxation. Ang-(1-7) also induced an increase of3H-arachidonic acid release and NO release from

Mas-transfected cells that were blocked by the specific Ang-(1-7) receptor Mas antagonist, A-779, but not by AT1or

AT2antagonists.21,22

Received November 30, 2005; first decision December 24, 2005; revision accepted February 14, 2006.

From the Departments of Physiology and Biophysics (R.A.S.S., C.H.C., A.P.A., R.D.d.P.), Morphology (E.G., G.T.K., A.J.F.), and Biochemistry and Immunology (J.S.C., A.S.R.), Biological Sciences Institute, Belo Horizonte, MG, Brazil; Department of Pediatrics (S.V.B.P.), Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Hypertension Unit (K.T.R., M.C.I.), Heart Institute, University of Sao Paulo, Sao Paulo, Brazil; and Max-Delbru¨ck-Center for Molecular Medicine (M.B., N.A.), Berlin-Buch, Germany.

Correspondence to Robson A.S. Santos, Departments of Physiology and Biophysics, Av. Antoˆnio Carlos, 6627 Biological Sciences Institute, UFMG, 31 270-901 Belo Horizonte, MG, Brazil. E-mail marrob@ciclope.Lcc.ufmg.br

© 2006 American Heart Association, Inc.

Hypertensionis available at http://www.hypertensionaha.org DOI: 10.1161/01.HYP.0000215289.51180.5c 996

(2)

However, there are little data available concerning the poten-tial role of Mas in the heart. Thus, the aim of this study was to investigate the effects of genetic deletion of Mas on cardiac function and coronary vascular resistance. In addition, the effect of genetic deletion of Mas on the expression of extracellular matrix protein in the heart was also investigated.

Methods

An expanded Methods section detailing the techniques and proce-dures mentioned here is available in an online supplement at http://www.hypertensionaha.org.

Mas-Knockout Mice

Wild-type (Mas⫹/⫹) and Mas knockout (Mas⫺/⫺)24 C57BL/6 mice were obtained from the transgenic animal facilities of the Laboratory of Hypertension, Federal University of Minas Gerais. All of the experimental protocols were performed in accordance with the guidelines for the humane use of laboratory animals established at our institution.

In Vitro Fluorescent–Labeled Ang-(1-7) Binding

Hearts from 3-month– old Mas⫹/⫹ (n3) and Mas⫺/⫺ (n2) mice

were snap-frozen in cooled isopentane. Cryostat sections (6␮m) were serially cut, mounted on gelatin-coated slides, and dried at 4°C, before the assay. The slices were preincubated in the assay buffer (10⫺2

M Na-phosphate buffer, pH 7.4; 1.2⫻10⫺1

M NaCl; 5 mmol/L MgCl2; 0.2% BSA; and 0.005% bacitracin) for 30 minutes. The experiments were performed using assay buffer containing 2.5⫻10⫺5 M phenylmethylsulfonyl fluoride; 6⫻10⫺4

M 1,10-phenanthroline; and 2⫻10⫺9M rhodamine-labeled Ang-(1-7) in the presence (non-specific binding) or absence ((non-specific binding) of Ang-(1-7) (10⫺6 mol/L) for 1 hour at 22°C to 24°C. Relative fluorescence measure-ments were performed on a Zeiss 510 Meta confocal microscope using a⫻63 oil-immersion objective lens.

Isolated Mouse Heart Technique

Seven male Mas⫹/⫹and 4 Mas⫺/⫺mice (3 months old) were used for

isolated hearts experiments as described previously.11

Echocardiography Studies

Echocardiographic features were obtained using the recommenda-tions of American Society of Echocardiography. All of the transtho-racic echocardiograms were performed in male Mas⫹/⫹and Mas⫺/⫺

mice (6 months old; n⫽10) by a single, blinded observer with the use of a Sequoia 512 (ACUSON Corp), which offers a 10- to 14-MHz multifrequencial linear transducer.

Electrophysiological Analysis

Individual mouse ventricular myocytes (n⫽8 cardiomyocytes from 3 different animals) were isolated from Mas⫹/⫹and Mas⫺/⫺animals

from the same littermates at 3 months of age. All of the experiments were performed using conventional whole-cell patch-clamp tech-niques at room temperature.25,26

Immunohistochemical Analysis

Immunofluorescence labeling and quantitative confocal microscopy were used to investigate the distribution and quantity of collagen types I, III, and VI and fibronectin present in Mas⫹/⫹ and Mas⫺/⫺

mice hearts.

from the BP measurement) were recorded on a data acquisition system (MP 100, Biopac Systems).

Statistical Analysis

Data are reported as mean⫾SEM. Statistical analyses for the binding assay, echocardiography, electrophysiological studies, and BP mea-surements were performed using unpaired Studentttest. Immuno-histochemistry statistical analysis was performed using unpaired Student t test followed by the Mann Whitney test. Statistical significance for isolated perfused heart experiments was estimated using 2-way ANOVA followed by Bonferroni test. Pⱕ0.05 was considered significant.

Results

Fluorescent-labeled Ang-(1-7) binding was found in the cardiomyocytes of Mas⫹/⫹ mice as displayed by confocal

analysis (Figure 1). Figure 1A shows total binding in Mas⫹/⫹

cardiomyocytes, which presented a punctuated staining pat-tern as would be expected for a G protein– coupled receptor. Conversely, specific binding of rhodamine-labeled Ang-(1-7) to heart slices from Mas⫺/⫺evaluated by quantitative confocal

microscopy was essentially abolished (Figure 1E).

Figure 1.In vitro localization of rhodamine–Ang-(1-7) binding in mice left ventricle slices. Figure shows total binding in Mas⫹/⫹

cardiomyocytes (A), nonspecific binding in Mas⫹/⫹

cardiomyo-cytes (B), total binding in Mas⫺/⫺cardiomyocytes (C), and

non-specific binding in Mas⫺/⫺cardiomyocytes (D). Concentration of

rhodamine-Ang-(1-7) was 2⫻10⫺9mol/L in the presence

(non-specific binding) or absence ((non-specific binding) of Ang-(1-7) (10⫺6

mol/L). Quantification of relative fluorescence in Mas⫹/⫹and

Mas⫺/⫺cardiomyocytes (E). Data are shown as the SD. *P0.01

(3)

Mas⫺/⫺ mice presented an impairment of in vitro heart

function. As shown in Figure 2A, systolic tension was significantly lower in hearts from Mas⫺/⫺ mice (average:

1.4⫾0.09 versus 2.1⫾0.03 g in age-matched Mas⫹/⫹ mice;

P⬍0.0001). Diastolic tension was similar in both groups (Figure 2B). Mas-deficient mice presented a lower ⫹dT/dt and ⫺dT/dt (average: 49.5⫾1.3 versus 65.2⫾0.3 and 49.5⫾1.5 versus 64.2⫾1.0 g/s in age-matched Mas⫹/⫹mice,

P⬍0.0001, respectively; Figure 2C and 2D) and a lower HR (average: 250.8⫾9.5 versus 333.3⫾2.6 bpm in age-matched Mas⫹/⫹ mice; P0.0001; Figure 2E). In addition, isolated

hearts from Mas⫺/⫺ mice presented a significantly higher

coronary perfusion pressure (average of the 30-minute re-cording: 172.7⫾3.0 versus 126.9⫾2.8 mm Hg in age-matched Mas⫹/⫹mice;P0.001; Figure 2F).

The Table summarizes the echocardiographic parameters obtained in Mas⫹/⫹ and Mas⫺/⫺ mice. In agreement with the

results obtained with isolated hearts, intact hearts in Mas-deficient mice showed a significantly lower fractional shortening (19.01⫾0.71 versus 22.90⫾0.25% in Mas⫹/⫹mice;P0.001).

Morphologically, Mas⫺/⫺mice displayed a significantly lower

posterior wall thickness in systole (0.085⫾0.001 versus 0.090⫾0.001 mm in Mas⫹/⫹ mice;P0.05) and left ventricle

end-diastolic dimension (0.387⫾0.001 versus 0.395⫾0.003 cm in Mas⫹/⫹mice;P0.05) and a higher left ventricle end-systolic

dimension (0.313⫾0.002 versus 0.305⫾0.002 cm in Mas⫹/⫹

mice;P⬍0.01). A substantially lower global ventricular func-tion, expressed by a higher myocardial performance index, was also observed (0.73⫾0.05 versus 0.54⫾0.03 in Mas⫹/⫹

mice;P⬍0.01; Figure 3).

To elucidate the possible mechanisms underlying the cardiac function impairment, we analyzed the whole-cell L-type Ca2⫹current in individual cardiomyocytes and cardiac

expression of collagen types I, III, and VI and fibronectin. Although it is now generally accepted that L-type Ca2⫹

current is the major trigger for sarcoplasmic reticulum Ca2⫹

release, no significant differences were observed between L-type Ca2⫹ current density from age-matched Mas⫹/⫹ and

Mas⫺/⫺mice (Figure 4A through 4D). Interestingly, the time

to the peak of Ca2⫹current was statistically different when Figure 2.Time course of systolic tension (A), diastolic tension (B),⫹dT/dt (C), ⫺dT/dt (D), HR (E), and coronary perfu-sion pressure (F) in isolated perfused mouse heart. The hearts were perfused according to Langendorff technique. **P⬍0.001, ***P⬍0.0001 (2-way ANOVA followed by Bonferroni test).

(4)

comparing the 2 groups (8.5⫾0.4 ms for the Mas⫹/⫹versus

10.8⫾0.6 ms for the Mas⫺/⫺ at 10 mV; P0.01; and

7.3⫾0.6 ms for the Mas⫹/⫹versus 9.40.4 ms for the Mas⫺/⫺

at 0 mV; P⬍0.01). This change could contribute to the cardiac dysfunction described.

Strikingly, we observed that the expression of several matrix proteins were significantly and markedly higher in the ventricles of adult Mas⫺/⫺mice hearts compared with Mas⫹/⫹

control mice (Figure 5A and 5B): that is, type I collagen 94.22⫾4.90 versus 59.00⫾2.35 in Mas⫹/⫹mice (P0.0001);

type collagen III 111.20⫾8.09 versus 45.67⫾2.21 in Mas⫹/⫹

mice (P⬍0.001); and fibronectin 78.89⫾2.90 versus 45.67⫾2.21 in Mas⫹/⫹ mice (P0.0001). Conversely, the

expression of type VI collagen in Mas⫺/⫺mice was decreased

20.78⫾2.29 versus 43.22⫾3.29 in Mas⫹/⫹mice (P0.0001).

increased BP, an additional group of animals at the same age of those used for echocardiograph measurements (6 months) were instrumented for acute BP recordings. As expected,24,27 no

differences in BP between Mas⫺/⫺ and Mas⫹/⫹ mice were

observed (BP: 79.3⫾4.9 versus 90.3⫾6.4 mm Hg,P⫽0.2438; HR: 467.0⫾56.1 versus 511.3⫾24.2 bpm,P⫽0.5086 in age-matched Mas⫹/⫹ mice under halothane anesthesia and BP:

100.0⫾3.0 versus 104.3⫾2.9 mm Hg, P⫽0.3622; HR: 559.7⫾21.2 versus 613.3⫾27.2 bpm,P⫽0.1957 in Mas⫹/⫹mice

after recovery of anesthesia). Actually, halothane-anesthetized Mas⫺/⫺ animals presented slight lower BP in comparison to

Mas⫹/⫹, probably resulting from the lower cardiac output.

Discussion

In this study, we have unmasked a key functional role of the Ang-(1-7) receptor Mas in the heart. We observed that systolic tension, ⫹dT/dt, and ⫺dT/dt were significantly lower in isolated hearts of Mas-deficient mice. Accordingly, an impaired heart function was observed in vivo by means of echocardiography. Echocardiographic measurements re-vealed a decreased fractional shortening, posterior wall thick-ness in systole and left ventricle end-diastolic dimension, and a higher left ventricle end-systolic dimension. A markedly lower global ventricular function, as defined by a higher myocardial performance index, was observed. In addition, Mas⫺/⫺mice presented a higher coronary perfusion pressure

compared with Mas⫹/⫹mice. These findings are in keeping

with several studies suggesting that Ang-(1-7) has beneficial effects in myocardial function.8 –13,16,28We have shown that

Ang-(1-7), at subnanomolar concentration, has an antiar-rhythmogenic effect on reperfusion11and that it preserves the

cardiac function in the postischemic period12 in isolated

perfused rat hearts. These effects were completely blocked by the selective Ang-(1-7) receptor Mas antagonist A-779, suggesting that these Ang-(1-7) actions in the heart are mediated by its specific receptor. One possible mechanism that may contribute to the beneficial effects of Ang-(1-7) in the heart is the modulation of cardiac Ang II levels, as described recently by Mendes et al.29 The present results

indicate that the Ang-(1-7) receptor Mas mediates the effects of Ang-(1-7) on the cardiac function.

The higher perfusion pressure observed in the isolated heart preparation is in agreement with the previous reports of a vasorelaxant activity of Ang-(1-7) in the coronary bed.8,19

Brosnihan et al9found that this heptapeptide elicits a

vaso-dilator response in canine coronary arteries. This effect was completely blocked by the nonselective Ang antagonist, [Sar1Thr8]-Ang II, but not by either an AT

1receptor

antago-nist or an AT2 receptor antagonist, suggesting that the

Ang-(1-7)-induced vasodilation in coronary arteries is medi-ated by a non-AT1/AT2 receptor. Our data indicate that the

Ang-(1-7) receptor Mas may account for Ang-(1-7)-induced vasodilation in the coronary bed. Accordingly, the Mas

Posterior wall thickness in systole, mm 0.090⫾0.001 0.085⫾0.001† Interventricular septal thickness in diastole, mm 0.067⫾0.001 0.066⫾0.001 Interventricular septal thickness in systole, mm 0.089⫾0.001 0.084⫾0.001 Left ventricle end-diastolic

dimension, cm

0.395⫾0.003 0.387⫾0.001† Left ventricle end-systolic

dimension, cm

0.305⫾0.002 0.313⫾0.002‡ Myocardial performance index 0.54⫾0.03 0.73⫾0.05‡ Isovolumic relaxation time, ms 19.50⫾1.18 20.60⫾0.88 Isovolumic relaxation time/HR 1.80⫾0.10 1.88⫾0.07

Heart rate, bpm 512.9⫾9.4 501.8⫾10.5

Left ventricular mass, g 0.085⫾0.002 0.082⫾0.003 Values are mean⫾SEM. Transthoracic echocardiogram was performed in male Mas⫹/⫹and Mas⫺/⫺mice (n10 for each group). Data were analyzed by

unpaired Studentttest.

*P⬍0.0001; †P⬍0.05; ‡P⬍0.01.

Figure 3.Representative echocardiographic measurements of contracting hearts from Mas⫹/⫹and Mas⫺/⫺. LVESD indicates

left ventricle systolic dimension; LVEDD, left ventricle end-diastolic dimension.

(5)

receptor antagonist A-779 abolished the potentiating effect of Ang-(1-7) on bradykinin-induced vasodilation in an isolated perfused rat heart preparation30and the vasodilation induced

by Ang-(1-7) in losartan-treated isolated mouse heart.20This

later effect was also abolished in isolated hearts of Mas-deficient mice.20

Recent reports have suggested that Ang-(1-7) could be a modulator of ion channel functioning similar to Ang II.31–33

Both peptides augment the release of [3H] norepinephrine

elicited by nerve stimulation in isolated rat atria.31 At the

neuromuscular junction, Ang-(1-7) increased acetylcholine release in a dose-dependent manner.33Thus, the lower

con-tractility observed in Mas⫺/⫺ mice provides additional

evi-dence that interactions between Ang-(1-7) and Mas receptor play an important modulatory role in calcium homeostasis in the heart tissue. Our data also indicate that the time to the peak of Ca2⫹ current measured at 10 and 0 mV for the

L-type Ca2⫹current was slower for the Mas⫺/⫺cells, which

could impair the sarcoplasmic reticulum refilling process and, thus, lead to less Ca2⫹being released during excitation.

We have suggested that Ang-(1-7) could produce changes in pacemaker ionic currents. Isolated hearts from transgenic rats [TGR(A1-7)3292] that chronically overproduce Ang-(1-7) resulting in 2.5-fold increase in circulating Ang-Ang-(1-7) concentration showed a significantly higher basal intrinsic HR observed in vivo and in ex vivo conditions.16On the other

hand, in Mas⫺/⫺mice, the basal intrinsic HR was significantly

lower compared with Mas⫹/⫹mice. In line with these data, De

Mello14reported that Ang-(1-7) activates the sodium pump,

hyperpolarizes the heart cell, and re-establishes the impulse conduction during ischemia/reperfusion, indicating that the antiarrhythmogenic effect of Ang-(1-7) could be because of changes in ionic currents. These observations suggest that Ang-(1-7) could actually modulate some component of the pacemaker ionic currents through Ang-(1-7) Mas receptor

stimulation. However, the exact mechanism involved in the change in HR in Mas-deficient mice remains to be established.

The changes in time to the peak of Ca2⫹current could be

related to the actual amount of Ca2⫹that is mobilized during

an action potential, but because we did not measure the global Ca2⫹transients elicited by electrical stimulation, we cannot

address this point. However, a major mechanism for the impairment in heart function in Mas⫺/⫺ appears to be the

strikingly changes in extracellular matrix proteins. Our results provide strong evidence for a previously unexpected key role for the Ang-(1-7)-Mas axis in the control of expression of extracellular matrix proteins. The fact that collagen VI undergoes an opposite change as that observed for collagen I, III, and fibronectin suggests that this influence is selective. These observations are in accordance with a recent report by Iwata et al,17 showing that Ang-(1-7) produces antifibrotic

effects. As expected,24,27a contribution of BP to the changes

in collagen expression or in cardiac function could be excluded by the direct BP measurements in halothane-anes-thetized and in nonaneshalothane-anes-thetized mice. Actually, under halo-thane anesthesia, the mean arterial pressure of Mas⫺/⫺mice

was slightly lower than that of Mas⫹/⫹mice, probably because

of the lower cardiac output. Because the expression of extracellular matrix proteins presented a similar pattern in neonate mice, we can further exclude a contribution of BP or systemic vascular resistance in Mas⫺/⫺as a primary cause for

the cardiac phenotype observed. Furthermore, Walther et al24

showed that Mas⫺/⫺are healthy, grow normally, and show no

obvious developmental abnormalities. They also reported no difference in drinking behavior. It is well known that fibrosis leads to alterations in diastolic function. Accordingly, asso-ciated with the important deficit of systolic function, we have also found a significantly lower dimension of the left ventri-cle at the end of diastole and a decreased⫺dT/dt in isolated

Figure 4.Electrophysiological analysis. Representative L-type Ca2⫹currents in

ventricular cardiomyocytes from Mas⫹/⫹

(A) and Mas⫺/⫺(B). (C) voltage

depen-dence ofIca,L, plotted as current density

(A/F) obtained for test potentials from ⫺50 to⫹60 mV. Values represent mean ⫾SEM. (D) membrane capacitance (pF) measured in Mas⫹/⫹() and Mas⫺/⫺(f) ventricular myocytes. Bar graph values are 157.5⫾12.9 (Mas⫹/⫹, n8) and

(6)

hearts of Mas⫺/⫺. Walther et al27have reported an increased

BP variability and indirect evidence for an increased sympa-thetic tonus in male Mas⫺/⫺mice as compared with control

mice (presumably C57BL/6 mice) using spectral analysis of BP data sampled at a relatively low frequency (250 Hz). Whether these changes contribute or are consequence or adaptative responses to the lower cardiac function remains to be established. Taken all together, these data indicate a BP-independent fibrosis as the main cause of heart dysfunc-tion in Mas-deficient mice.

One of the limitations of our study is related to the echocardiogram measurements. Reporting of E and A peaks and E/A relation, as well as E deceleration time would add an important information. However, because the halothane an-esthetic regimen keeps HR near physiological levels and the HR of mice is very high, peak E fuses with peak A. Besides, values of fused peak E and A did not differ between the groups, in accordance with isovolumic relaxation time.

important concern. In this study, halothane has been used to perform echocardiogram, because it is more convenient and reliable with respect to rate of induction, reversibility, and control of anesthetic depth.34In addition, HR is less affected

with inhaled anesthetic than with intraperitoneal ones. Al-though further studies with other anesthetics, such as isoflu-rane, would be interesting to confirm our findings with halothane, the similarity between our in vivo and in vitro data provides a very strong evidence for a key role of the Ang-(1-7)-Mas axis in heart function.

Perspectives

The changes in extracellular matrix proteins to a profibrotic profile in Mas⫺/⫺ mice provide additional evidence that the

Ang-(1-7)-Mas axis plays a key role in the beneficial effects of angiotensin-converting enzyme (ACE) inhibitors, AT1

receptor blockers (ARBs), and aldosterone antagonists in the heart. It has been shown that chronic treatment with ACE inhibitors or ARBs increases the activity of Ang-(1-7)– forming enzymes,35–37 in particular, heart ACE2.36 More

recently, it has been demonstrated in humans that treatment with aldosterone antagonists substantially increased ACE2 activity.38 The prominent decrease in cardiac function in

Mas⫺/⫺ mice, which can be correlated with the marked

changes in extracellular matrix proteins and the antihypertro-phic effects of Ang-(1-7),16,17 apparently mediated by its

interaction with Mas,18suggest that activation of the

Ang-(1-7)–forming pathways by RAS blockade produces beneficial cardiac effects through activation of Mas. One could argue that ACE2 knockout mice39showed more pronounced cardiac

function impairment than Mas knockout mice. However, the ACE2 knockout mice presented a higher Ang II level, which may contribute to the cardiac phenotype. Of note is the fact that, in ACE2 knockout mice, the cardiac dysfunctions appeared only after 6 months of age, whereas the cardiac fibrotic phenotype in Mas⫺/⫺ mice was already observed in

the neonatal period.

In conclusion, our findings suggest that Ang-(1-7)-Mas axis plays a key role in the heart. Furthermore, our data also suggest that this RAS axis may be a potential target for the development of new cardiovascular drugs.7

Acknowledgments

This work was supported in part by Conselho Nacional de Desen-volvimento Cientı´fico e Tecnolo´gico–Programa de Grupos de Exce-leˆncia and Fundac¸a˜o de Amparo a` Pesquisa de Minas Gerais. We are thankful to Jose´ Roberto da Silva. The Zeiss confocal microscope is located in the Centro de Microscopia, Instituto de Cieˆncias Biolo´gi-cas, Universidade Federal de Minas Gerais.

References

1. Atlas SA. The renin-angiotensin system revisited: classical and non-classical pathways of angiotensin formation.Mt Sinai J Med. 1998;65: 87–96.

Figure 5.Immunofluorescent localization of matrix proteins in the left ventricles of adult Mas⫹/⫹and Mas⫺/⫺mice hearts.

Expression of type I collagen, type III collagen, and fibronectin were increased in the left ventricles of Mas⫺/⫺compared with

Mas⫹/⫹mice, whereas the expression of type VI collagen was

decreased. The images in the control column show the level of immunostaining obtained when the primary antibody is omitted from the incubation procedure (A). Quantification of extracellular matrix proteins in the left ventricles of Mas⫹/⫹and Mas⫺/⫺mice

(B). Data are shown as the SEM. *P⬍0.001; **P⬍0.0001. A.U. indicates arbitrary unit.

(7)

2. Ardaillou R, Michel JB. The relative roles of circulating and tissue renin-angiotensin systems.Nephrol Dial Transplant. 1999;14:283–286. 3. Oudit GY, Crackower MA, Backx PH, Penninger JM. The role of ACE2

in cadiovascular physiology.Trends Cardiovasc Med. 2003;13:93–101. 4. Santos RAS, Campagnole-Santos MJ, Andrade SP. Angiotensin-(1-7): an

update.Regul Pept. 2000;91:45– 62.

5. Reudelhuber TL. The renin-angiotensin system: peptides and enzymes beyond angiotensin II.Curr Opin Nephrol Hypertens. 2005;14:155–159. 6. Ferrario CM, Trask AJ, Jessup JA. Advances in the biochemical and functional roles of angiotensin converting enzyme 2 and angioten-sin-(1-7) in the regulation of cardiovascular function.Am J Physiol Heart Circ Physiol. 2005;289:H2281–H2290.

7. Santos RAS, Ferreira AJ, Pinheiro SV, Sampaio WO, Touyz R, Campagnole-Santos MJ. Angiotensin-(1-7) and its receptor as a potential targets for new cardiovascular drugs.Expert Opin Investig Drugs. 2005; 14:1019 –1031.

8. Brosnihan KB, Li P, Ferrario CM. Angiotensin-(1-7) dilates canine coronary arteries through kinins and nitric oxide.Hypertension. 1996;27: 523–528.

9. Brosnihan KB, Li P, Tallant EA, Ferrario CM. Angiotensin-(1-7): a novel vasodilator of the coronary circulation.Biol Res. 1998;31:227–234. 10. Loot AE, Roks AJM, Henning RH, Tio RA, Suurmeijer AJH, Boomsma

F, van Gilst WH. Angiotensin-(1-7) attenuates the development of heart failure after myocardial infarction in rats. Circulation. 2002;105: 1548 –1550.

11. Ferreira AJ, Santos RAS, Almeida AP. Angiotensin-(1-7): cardiopro-tective effect in myocardial ischemia/reperfusion.Hypertension. 2001;38: 665– 668.

12. Ferreira AJ, Santos RAS, Almeida AP. Angiotensin-(1-7) improves the post-ischemic function in isolated perfused rat hearts.Braz J Med Biol Res. 2002;35:1083–1090.

13. Averill DB, Ishiyama Y, Chappell MC, Ferrario CM. Cardiac angioten-sin-(1-7) in ischemic cardiomyopathy.Circulation. 2003;108:2141–2146. 14. De Mello WC. Angiotensin (1-7) re-establishes impulse conduction in cardiac muscle during ischaemia-reperfusion. The role of the sodium pump.J Renin Angiotensin Aldosterone Syst. 2004;5:203–208. 15. Sampaio WO, Nascimento AA, Santos RAS. Systemic and regional

hemodynamic effects of angiotensin-(1-7) in rats.Am J Physiol Heart Circ Physiol. 2003;284:H1985–H1994.

16. Santos RAS, Ferreira AJ, Nadu AP, Braga AN, Almeida AP, Campagnole-Santos MJ, Baltatu O, Iliescu R, Reudelhuber TL, Bader M. Expression of an angiotensin-(1-7)-producing fusion protein produces cardioprotective effects in rats.Physiol Genomics. 2004;17:292–299. 17. Iwata M, Cowling RT, Gurantz D, Moore C, Zhang S, Yuan JX,

Greenberg BH. Angiotensin-(1-7) binds to specific receptors on cardiac fibroblasts to initiate anti-fibrotic and anti-trophic effects.Am J Physiol Heart Circ Physiol. 2005;289:H2356 –H2363.

18. Tallant EA, Ferrario CM, Gallagher PE. Angiotensin-(1-7) inhibits growth of cardiac myocytes through activation of the mas receptor.Am J Physiol Heart Circ Physiol. 2005;289:H1560 –H1566.

19. Porsti I, Bara AT, Busse R, Hecker M. Release of nitric oxide by angiotensin-(1-7) from porcine coronary endothelium: implications for a novel angiotensin receptor.Br J Pharmacol. 1994;111:652– 654. 20. Castro CH, Santos RAS, Ferreira AJ, Bader M, Alenina N, Almeida AP.

Evidence for a functional interaction of the angiotensin-(1-7) receptor Mas with AT1 and AT2 receptors in the mouse heart.Hypertension. 2005;46:937–942.

21. Santos RAS, Simo˜es e Silva AC, Maric C, Silva DM, Machado RP, Buhr I, Heringer-Walther S, Pinheiro SV, Lopes MT, Bader M, Mendes EP, Lemos VS, Campagnole-Santos MJ, Schultheiss HP, Speth R, Walther T. Angiotensin-(1-7) is an endogenous ligand for the G protein-coupled receptorMas. Proc Natl Acad Sci U S A. 2003;100:8258 – 8263.

22. Pinheiro SV, Simo˜es e Silva AC, Sampaio WO, Paula RD, Mendes EP, Bontempo ED, Pesquero JB, Walther T, Alenina N, Bader M, Bleich M, Santos RAS. Nonpeptide AVE 0991 is an angiotensin-(1-7) receptor Mas agonist in the mouse kidney.Hypertension. 2004;44:490 – 496. 23. Metzger R, Bader M, Ludwig T, Berberich C, Bunnemann B, Ganten D.

Expression of the mouse and rat mas proto-oncogene in the brain and peripheral tissues.FEBS Lett. 1995;357:27–32.

24. Walther T, Balschun D, Voigt JP, Fink H, Zuschratter W, Birchmeier C, Ganten D, Bader M. Sustained long term potentiation and anxiety in mice lacking the Mas protooncogene.J Biol Chem. 1998;273:11867–11873. 25. Hamill OP, Marty A, Neher E, Sakmann B, Sigworth FJ. Improved

patch-clamp techniques for high-resolution current recording from cells and cell-free membrane patches.Pflugers Arch. 1981;391:85–100. 26. Guatimosim S, Sobie EA, Cruz JS, Martin LA, Lederer WJ. Molecular

identification of a TTX-sensitive Ca2⫹current.Am J Physiol Cell Physiol.

2001;280:C1327–C1339.

27. Walther T, Wessel N, Kang N, Sander A, Tschope C, Malberg H, Bader M, Voss A. Altered heart rate and blood pressure variability in mice lacking the Mas protooncogene.Braz J Med Biol Res. 2000;33:1–9. 28. Zisman LS, Keller RS, Weaver B, Lin Q, Speth R, Bristow MR, Canver

CC. Increased angiotensin-(1-7)-forming activity in failing human heart ventricles: evidence for upregulation of the angiotensin-converting enzyme homologue ACE2.Circulation. 2003;108:1707–1712. 29. Mendes AC, Ferreira AJ, Pinheiro SV, Santos RAS. Chronic infusion of

angiotensin-(1-7) reduces heart angiotensin II levels in rats.Regul Pept. 2005;125:29 –34.

30. Almeida AP, Fra´bregas BC, Madureira MM, Santos RJS, Campagnole-Santos MJ, Campagnole-Santos RAS. Angiotensin-(1-7) potentiates the coronary vaso-dilatatory effect of bradykinin in the isolated rat heart.Braz J Med Biol Res. 2000;33:709 –713.

31. Gironacci MM, Adler-Graschinsky E, Pena C, Enero MA. Effects of angiotensin II and angiotensin-(1-7) on the release of [3H]norepinephrine

from rat atria.Hypertension. 1994;24:457– 460.

32. Gironacci MM, Yujnovsky I, Gorzalczany S, Taira C, Pena C. Angio-tensin-(1-7) inhibits the angiotensin II-enhanced norepinephrine release in coarcted hypertensive rats.Regul Pept. 2004;118:45– 49.

33. Bevilaqua ER, Kushmerick C, Beira˜o PSL, Naves LA. Angiotensin-(1-7) increases quantal content and facilitation at the frog neuromuscular junction.Brain Res. 2002;927:208 –211.

34. Chaves AA, Weinstein DM, Bauer JA. Non-invasive echocardiographic studies in mice: Influence of anesthetic regimen. Life Sci. 2001;69: 213–222.

35. Stanziola L, Greene LJ, Santos RAS. Effect of chronic angiotensin con-verting enzyme inhibition on angiotensin I and bradykinin metabolism in rats.Am J Hypertens. 1999;12:1021–1029.

36. Ishiyama Y, Gallagher PE, Averill DB, Tallant EA, Brosnihan KB, Ferrario CM. Upregulation of angiotensin-converting enzyme 2 after myocardial infarction by blockade of angiotensin II receptors. Hyper-tension. 2004;43:970 –976.

37. Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, Diz DI, Gallagher PE. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2.Circulation. 2005;111:2605–2610.

38. Keidar S, Gamliel-Lazarovich A, Kaplan M, Pavlotzky E, Hamoud S, Hayek T, Karry R, Abassi Z. Mineralocorticoid receptor blocker increases angiotensin-converting enzyme 2 activity in congestive heart failure patients.Circ Res. 2005;97:946 –953.

39. Crackower MA, Sarao R, Oudit GY, Yagil C, Kozieradzki I, Scanga SE, Oliveira-dos-Santos AJ, da Costa J, Zhang L, Pei Y, Scholey J, Ferrario CM, Manoukian AS, Chappell MC, Backx PH, Yagil Y, Penninger JM. Angiotensin-converting enzyme 2 is an essential regulator of heart function.Nature. 2002;417:822– 828.

References

Related documents

[78], presented in this literature a control strategy is proposed to regulate the voltage across the FCs at their respective reference voltage levels by swapping the switching

Firstly, At P15, in Arg1 KO mice, the density of myelinated axons was significantly reduced in subcortical white matter of the motor cortex, pyramidal tract in medulla, and CST in

The spatial analysis revealed that the area with the high- est probability of hosting a new subaerial eruption is mainly located in the same area than the previous 1824 and Timan-

It was decided that with the presence of such significant red flag signs that she should undergo advanced imaging, in this case an MRI, that revealed an underlying malignancy, which

He also spreads, south dakota goose guides over a entire season hunts included with your south dakota hunting in south dakota snows while attending university of canada. Done out

Therefore, many efforts have been devoted to solve most optimal Job Shop Scheduling Problems (JSSP), as most of the researches aimed at minimizing the maximum completion time. JSSP

evaluate the clinical course of DVT. As a main objective, we compared the effects of two therapeutic options for DVT, both of which were used as a standard regimen at

The center is dedicated to developing and supporting high quality mathematics programs that interface with the K-16 community, including outreach programs for K-12 students,