Reduced isoproterenol-induced renin-angiotensin changes and extracellular matrix deposition in...

8
Research Article Reduced isoproterenol-induced renin-angiotensin changes and extracellular matrix deposition in hearts of TGR(A1–7)3292 rats Ana Paula Nadu, PhD a , Anderson J. Ferreira, PhD b , Timothy L. Reudelhuber, PhD c , Michael Bader, PhD d , and Robson A. S. Santos, MD a, * a Departments of Physiology and Biophysics and b Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; c Laboratory of Molecular Biochemistry of Hypertension, Clinical Research Institute of Montreal, Quebec, Canada; and d Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany Manuscript received February 24, 2008 and accepted April 17, 2008 Abstract We investigated the expression of specific extracellular matrix (ECM) proteins in cardiac hypertrophy induced by isoproterenol in TGR(A1–7)3292 rats. Additionally, changes in circulating and tissue renin-angiotensin system (RAS) were analyzed. Left ventricles (LV) were used for quantification of collagen type I, III, and fibronectin using immunofluorescence-labeling techniques. Angiotensin (Ang) II levels were measured by radioimmunoassay. Expression of RAS components was assessed by semi-quantitative poly- merase chain reaction (PCR) or real-time PCR. Isoproterenol treatment induced an increase in the expression of collagen I, III, and fibronectin in normal rats. Collagen I and fibronectin expression were decreased in TGR(A1–7)3292 at basal conditions and both proteins increased by isoproterenol treatment; however, the levels achieved were still significantly lower than those observed in treated normal rats. The increase in collagen III observed in normal rats was completely blunted in TGR(A1–7)3292. Moreover, TGR(A1–7)3292 presented lower Ang II levels and angiotensinogen expression and a higher angiotensin-converting enzyme 2 (ACE2) expression in LV. Isoproterenol treatment increased cardiac Ang II concentration only in normal rats, which was associated with an increase in ACE2 and a decrease in Mas expression. These observations suggest that Ang-(1–7) specifically modulates the expression of RAS components and ECM proteins in LV. J Am Soc Hypertens 2008;2(5): 341–348. © 2008 American Society of Hypertension. All rights reserved. Keywords: Mas receptor; ACE2; collagen; hypertrophy. Introduction Angiotensin (Ang) II, the main end-product of the renin- angiotensin system (RAS) cascade, stimulates the biosyn- thesis of cardiac extracellular matrix (ECM) proteins, lead- ing to interstitial and perivascular fibrosis. 1,2 On the other hand, Ang-(1–7), a bioactive component of the RAS, is believed to be an antifibrotic peptide. 3,4 Therefore, the amount of ECM proteins in the heart could also depend of the balance between Ang II and Ang-(1–7) levels. Many studies have indicated that the heart is an im- portant target for Ang-(1–7) actions. 5–7 Among its ben- eficial effects in the heart, the antifibrotic/antitrophic effect is believed to be one of the most important car- dioprotective actions of this peptide. The antiprolifera- tive and antitrophic properties of the Ang-(1–7) could be the mechanism by which this peptide improves the car- diac function and protects the heart against remodel- ing. 5,7–9 Ang-(1–7) reduces the growth of cardiomyo- cytes, 10 acting through the Mas receptor 11 and inhibits collagen synthesis in cardiac fibroblasts. 12 Hearts from Mas-deficient mice exhibits a marked change in ECM protein expression to a pro-fibrotic profile and important cardiac dysfunction. 3,13 In addition, it has been reported that Ang-(1–7) prevents cardiac fibrosis elicited by either This work was supported by CNPq-PRONEX (Conselho Na- cional de Desenvolvimento Científico e Tecnológico), FAPEMIG- PRONEX (Fundação de Amparo à Pesquisa de Minas Gerais), and CAPES (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior). Conflict of interest: none. *Corresponding author: Robson A. S. Santos, MD, Departa- mento de Fisiologia e Biofísica, Av. Antônio Carlos, 6627 – ICB – UFMG, 31.270-901 – Belo Horizonte, MG, Brazil. Tel: (55-31) 3499 2957; fax: (55-31) 3499 2924. E-mail: [email protected] Journal of the American Society of Hypertension 2(5) (2008) 341–348 1933-1711/08/$ – see front matter © 2008 American Society of Hypertension. All rights reserved. doi:10.1016/j.jash.2008.04.012

Transcript of Reduced isoproterenol-induced renin-angiotensin changes and extracellular matrix deposition in...

A

WT((mfiptT(weHK

I

atih

cPCS

m–3

1d

Research Article

Reduced isoproterenol-induced renin-angiotensin changes andextracellular matrix deposition in hearts of TGR(A1–7)3292 ratsAna Paula Nadu, PhDa, Anderson J. Ferreira, PhDb, Timothy L. Reudelhuber, PhDc,

Michael Bader, PhDd, and Robson A. S. Santos, MDa,*aDepartments of Physiology and Biophysics and

bMorphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil;cLaboratory of Molecular Biochemistry of Hypertension, Clinical Research Institute of Montreal, Quebec, Canada; and

dMax-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany

Manuscript received February 24, 2008 and accepted April 17, 2008

bstract

e investigated the expression of specific extracellular matrix (ECM) proteins in cardiac hypertrophy induced by isoproterenol inGR(A1–7)3292 rats. Additionally, changes in circulating and tissue renin-angiotensin system (RAS) were analyzed. Left ventricles

LV) were used for quantification of collagen type I, III, and fibronectin using immunofluorescence-labeling techniques. AngiotensinAng) II levels were measured by radioimmunoassay. Expression of RAS components was assessed by semi-quantitative poly-erase chain reaction (PCR) or real-time PCR. Isoproterenol treatment induced an increase in the expression of collagen I, III, andbronectin in normal rats. Collagen I and fibronectin expression were decreased in TGR(A1–7)3292 at basal conditions and bothroteins increased by isoproterenol treatment; however, the levels achieved were still significantly lower than those observed inreated normal rats. The increase in collagen III observed in normal rats was completely blunted in TGR(A1–7)3292. Moreover,GR(A1–7)3292 presented lower Ang II levels and angiotensinogen expression and a higher angiotensin-converting enzyme 2

ACE2) expression in LV. Isoproterenol treatment increased cardiac Ang II concentration only in normal rats, which was associatedith an increase in ACE2 and a decrease in Mas expression. These observations suggest that Ang-(1–7) specifically modulates the

xpression of RAS components and ECM proteins in LV. J Am Soc Hypertens 2008;2(5): 341–348. © 2008 American Society ofypertension. All rights reserved.

Journal of the American Society of Hypertension 2(5) (2008) 341–348

eywords: Mas receptor; ACE2; collagen; hypertrophy.

bat

peedttdiccMpc

ntroduction

Angiotensin (Ang) II, the main end-product of the renin-ngiotensin system (RAS) cascade, stimulates the biosyn-hesis of cardiac extracellular matrix (ECM) proteins, lead-ng to interstitial and perivascular fibrosis.1,2 On the otherand, Ang-(1–7), a bioactive component of the RAS, is

This work was supported by CNPq-PRONEX (Conselho Na-ional de Desenvolvimento Científico e Tecnológico), FAPEMIG-RONEX (Fundação de Amparo à Pesquisa de Minas Gerais), andAPES (Coordenação de Aperfeiçoamento de Pessoal de Níveluperior).

Conflict of interest: none.*Corresponding author: Robson A. S. Santos, MD, Departa-

ento de Fisiologia e Biofísica, Av. Antônio Carlos, 6627 – ICBUFMG, 31.270-901 – Belo Horizonte, MG, Brazil. Tel: (55-31)

499 2957; fax: (55-31) 3499 2924.

tE-mail: [email protected]

933-1711/08/$ – see front matter © 2008 American Society of Hyperteoi:10.1016/j.jash.2008.04.012

elieved to be an antifibrotic peptide.3,4 Therefore, themount of ECM proteins in the heart could also depend ofhe balance between Ang II and Ang-(1–7) levels.

Many studies have indicated that the heart is an im-ortant target for Ang-(1–7) actions.5–7 Among its ben-ficial effects in the heart, the antifibrotic/antitrophicffect is believed to be one of the most important car-ioprotective actions of this peptide. The antiprolifera-ive and antitrophic properties of the Ang-(1–7) could behe mechanism by which this peptide improves the car-iac function and protects the heart against remodel-ng.5,7–9 Ang-(1–7) reduces the growth of cardiomyo-ytes,10 acting through the Mas receptor11 and inhibitsollagen synthesis in cardiac fibroblasts.12 Hearts fromas-deficient mice exhibits a marked change in ECM

rotein expression to a pro-fibrotic profile and importantardiac dysfunction.3,13 In addition, it has been reported

hat Ang-(1–7) prevents cardiac fibrosis elicited by either

nsion. All rights reserved.

dAc

(blefetdrtTER

M

A

waUtta

H

ddvAttE

I

tpcdagal(

A

i

dedl�AammpmpIcbct

S

niCsDws((st3uCGACp(T5art

R

pC5puC

342 A.P. Nadu et al. / Journal of the American Society of Hypertension 2(5) (2008) 341–348

eoxycorticosterone acetate (DOCA)-salt treatment orng II infusion independent of blood pressure (BP)

hanges.6,14

We have found that a 2.5-fold increase in plasma Ang-1–7) levels in transgenic rats TGR(A1–7)3292 (TG) har-oring an Ang-(1–7)-producing fusion protein leads to aess pronounced cardiac hypertrophy induced by isoproter-nol, increase in dP/dt, reduction in the duration of reper-usion arrhythmias, and an improvement in the postisch-mic cardiac function in isolated perfused hearts.15 Also,his TG line presents an increased stroke volume and aecreased total peripheral resistance.16 The aim of the cur-ent study was to investigate the effects of isoproterenol onhe expression of specific ECM proteins in hearts fromGR(A1–7)3292 and determine whether the changes inCM expression are associated with alterations in cardiacAS components.

ethods

nimals

Male Sprague-Dawley (SD) and TGR(A1–7)3292 ratseighting 300 to 350 g were obtained from transgenic

nimal facilities at the Laboratory of Hypertension, Federalniversity of Minas Gerais, Brazil. All experimental pro-

ocols were performed in accordance with the guidelines forhe humane use of laboratory animals of our Institute andpproved by local authorities.

eart Hypertrophy

Heart hypertrophy was induced in SD and TG rats byaily injection of isoproterenol (2 mg/kg intraperitoneally, 7ays) and control groups (SD and TG) received injections ofehicle (0.9% NaCl 0.1 mL/100 g intraperitoneally, 7 days).t the end of the 7-day period, the rats were decapitated and

he hearts removed. The heart chambers were dissected andhe left ventricles (LV) were immediately frozen or fixed forCM protein analysis.

mmunohistochemical Analysis

The LVs of SD and TG rats treated or not with isopro-erenol (n � 3 for each group) were harvested, washed inhosphate-buffered saline to remove excess blood, andryofixed in a –80° C solution of 80% methanol and 20%imethyl sulfoxide for 7 days. Immunofluorescence labelingnd quantitative confocal microscopy were used to investi-ate the distribution and quantity of collagen types I, III,nd fibronectin according to Santos et al.3 Nuclei wereabeled with 4=6-diamidino-2-phenylindole dihydrochlorideDAPI) cat. #D1306, Molecular Probes (Carlsbad, CA).

ng II Measurements

After decapitation, the LVs of rats treated or not with

soproterenol (n � 6–9) were collected and frozen imme- C

iately. The organs were homogenized with 0.045 N HCl inthanol (10 mL/g of tissue) containing 0.9 �mol/L p-hy-roxymercuribenzoate, 131.5 �mol/L of 1,10-phenanthro-ine, 0.9 �mol/L phenylmethylsulfonyl fluoride, 1.75mol/L pepstatin A, 0.032% Ethylenediamine Tetraaceticcid (EDTA), and 0.0043% protease-free bovine serum

lbumin. In addition, blood samples for Ang II measure-ents were collected in polypropylene tubes containing 1mol/L p-hydroxymercuribenzoate, 30 mmol/L of 1,10-

henanthroline, 1 mmol/L phenylmethylsulfonyl fluoride, 1mol/L pepstatin A, and 7.5% EDTA. After extraction of

eptides using Bond-Elut phenylsilica cartridges (Varian,nc., Palo Alto, CA) from tissue homogenates and plasma,ollected as described above, Ang II levels were measuredy radioimmunoassay according to Botelho et al.17 Proteinoncentration in the crude homogenates was determined byhe Bradford method.18

emiquantitative PCR

LVs obtained from rats that received vehicle or isoprotere-ol (n � 3–4) were homogenized, and the total RNA wassolated with TRIzol reagent (Invitrogen Life Technologies,arlsbad, CA) according to the manufacturer’s protocol. RNA

amples (1 �g) were treated with DNAse to eliminate genomicNA present in the samples. Total RNA treated with DNAseas reverse transcribed using random primers. The single-

tranded cDNA was amplified by polymerase chain reactionPCR) using 33 cycles for angiotensin-converting enzyme 2ACE2) under the following conditions: 30 seconds, 95° C; 30econds, 60° C; 60 seconds, 72° C; and 35 cycles for angio-ensinogen under the following conditions: 30 seconds, 95° C;0 seconds, 56° C; 60 seconds, 72° C. cDNA was amplifiedsing the following set of primers: ACE2, forward 5=-AAAGTTCACTTGCTTCTTGG-3= and reverse 5=-TCAT-TAAATGGTGCTCATGG-3=; angiotensinogen, forward 5=-CACCCCTGCTACAGTCCAC-3= and reverse 5=-ACCC-CTCTAGTGGCAAGTT-3=. The same samples were em-loyed for Glyceraldehyde 3-phosphate dehydrogenaseGAPDH) cDNA amplification using the forward primer 5=-TGTTGCCATCAACGACCCC-3= and the reverse primer=-ATGAGCCCTTCCACAATGCC-3= to ensure that equalmounts of RNA samples were reversely transcribed. Theesults were reported as the ratio between ACE2 or angio-ensinogen and GAPDH values.

eal-Time PCR

Real-time PCR was used to assess Mas receptor ex-ression (n � 4 –5) using the forward primer 5=-TGAC-ATTGAACAGATTGCCA-3= and the reverse primer=-TGTAGTTTGTGACGGCTGGTG-3=. The same sam-les were employed for GAPDH cDNA amplificationsing the forward primer 5=-ATGTTCCAGTATGACTC-ACTCACG-3= and the reverse primer 5=-GAAGACAC-

AGTAGACTCCACGACA-3= to ensure that equal

aw

S

nfv

R

pbtlrwo

t0t2tp1�t

tpiAbai(t

343A.P. Nadu et al. / Journal of the American Society of Hypertension 2(5) (2008) 341–348

mounts of RNA were reversely transcribed. The resultsere reported as the ratio of Mas to GAPDH.

tatistical Analysis

All data are reported as means � SEM. Statistical sig-ificance was estimated using one-way analysis of varianceollowed by Newman-Keuls posttest or Student’s t test. A Palue � .05 was considered significant.

esults

Under basal conditions, TG rats presented a lower ex-ression of collagen type I and fibronectin (Figures 1 and 2),ut not of collagen type III in LVs (Figure 3). Isoproterenolreatment induced an increase in the expression of the col-agen type I, III, and fibronectin in SD rats. However, in TGats, only the expression of collagen type I and fibronectinere increased after isoproterenol treatment and the levels

f expression achieved were still significantly lower than v

hose observed in treated SD rats (collagen type I: 18.5 �.9 vs. 22.8 � 1.9 arbitrary units in SD, Figure 1; fibronec-in: 11.6 � 0.9 vs. 19.6 � 1.4 arbitrary units in SD, Figure). Interestingly, the increase in the expression of collagenype III observed in isoproterenol-treated SD rats was com-letely blunted in TG rats (TG: 13.60 � 1.07 vs. 13.64 �.04 arbitrary units in isoproterenol-treated rats; SD: 12.95

0.87 vs. 20.85 � 0.97 arbitrary units in isoproterenol-reated rats; Figure 3).

Because Ang II is closely involved in the regulation ofhe ECM proteins, its levels were evaluated in hearts andlasma of SD and TG rats at basal conditions and aftersoproterenol treatment. As shown in Table 1, cardiacng II levels were significantly decreased in TG rats atasal conditions and after isoproterenol treatment. Inddition, the increase in Ang II concentration induced bysoproterenol treatment observed in hearts from SD rats1.23 � 0.06 vs. 1.06 � 0.07 pg/mg protein in vehicle-reated SD rats), was not present in TG rats (0.83 � 0.10

Figure 1. Immunofluorescent localization of collagen type Iin LVs. Low-magnification images reveal the overall level ofimmunostaining in (A) SD vehicle, (B) SD ISO, (C)TGR(A1–7)3292 (TG) vehicle, and (D) TG ISO groups. (E)Negative control was obtained when the primary antibodywas omitted from the incubation procedure. Original mag-nification, 40�. (F) Quantification of collagen type I in LVs.Data are shown as means � SEM. A.U., arbitrary unit; ISO,isoproterenol; LV, left ventricle; SD, Sprague-Dawley. *P �.05. **P � .01 (one-way analysis of variance followed byNewman-Keuls posttest).

s. 0.77 � 0.04 pg/mg protein in vehicle-treated TG rats).

IrMt

taArSd(enebmti0(

D

wAdcAweTcsflo

apA

344 A.P. Nadu et al. / Journal of the American Society of Hypertension 2(5) (2008) 341–348

n contrast, plasma Ang II levels were augmented in TGats at basal conditions and after isoproterenol treatment.

oreover, isoproterenol administration did not changehe plasma Ang II concentration in SD and TG rats.

To evaluate possible mechanisms underlying the selec-ive change in cardiac Ang II levels observed in TG rats, wenalyzed the expression of ACE2 and angiotensinogen.CE2 expression was significantly increased in LVs of TG

ats at basal conditions (0.151 � 0.033 vs. 0.068 � 0.006 inD rats), but not after isoproterenol treatment, which in-uced an increase in ACE2 expression only in SD ratsTable 2). Hearts from TG rats also presented a lowerxpression of angiotensinogen before and after isoprotere-ol treatment. No significant changes in angiotensinogenxpression were observed after isoproterenol treatment inoth strains (Table 2). The expression of Mas receptorRNA was similar in SD and TG rats under basal condi-

ions. Isoproterenol treatment induced a significant decreasen the expression of Mas in SD rats (0.140 � 0.067 vs..541 � 0.140 in vehicle-treated rats), but not in TG rats

Figure 2. Immunofluorescent localization of fibronectin inLVs. Low-magnification images reveal the overall level ofimmunostaining in (A) SD vehicle, (B) SD ISO, (C)TGR(A1–7)3292 (TG) vehicle, and (D) TG ISO groups. (E)Negative control was obtained when the primary antibodywas omitted from the incubation procedure. Blue stainingcorresponds to nuclei labeled with 4=6-DAPI. Original mag-nification, 40�. (F) Quantification of fibronectin in LVs.Data are shown as means � SEM. A.U., arbitrary unit;DAPI, diamidino-2-phenylindole dihydrochloride; ISO, iso-proterenol; LV, left ventricle; SD, Sprague-Dawley. *P �.05. ***P � .001 (one-way analysis of variance followed byNewman-Keuls posttest).

Table 2). t

iscussion

In the current study, using a transgenic animal model,hich presents a suitable increase of 2.5-fold in the plasmang-(1–7) levels, we demonstrated that Ang-(1–7) plays aifferential control in the amount of specific ECM proteins,ontributing to the antifibrotic effect of this peptide. Theseng-(1–7) actions were further evidenced when the ratsere challenged with isoproterenol, a well-known �-adren-

rgic agonist able to induce cardiac remodeling. In addition,GR(A1–7)3292 rats showed a significant reduction in theardiac Ang II levels even after isoproterenol treatment,uggesting that Ang-(1–7) might exerts its antifibrotic ef-ects also through its regulating effect on the cardiac Ang IIevels, since this peptide is believed to induce accumulationf ECM proteins in the heart.Many studies have proposed that the cardioprotective

xis of the RAS composed by ACE2, Ang-(1–7), and Maslays antiproliferative effects in the heart.3,6,10,12,14,19,20

ng-(1–7) binds to a specific site distinct from Ang II

ype 1 and type 2 receptors in adult rat cardiac fibroblasts

t(soatoptAbicserpnipit

itdemfhlta

aTtMpacApe

345A.P. Nadu et al. / Journal of the American Society of Hypertension 2(5) (2008) 341–348

o inhibit the collagen synthesis.12 Interestingly, Ang-1–7) infusion prevents collagen deposition in DOCA-alt animals14 and in Ang II-infused rats (6) independentf changes in BP. In addition, the compound AVE 0991,synthetic Ang-(1–7) analogue, decreases the ECM pro-

ein deposition induced by isoproterenol treatment with-ut any changes in BP.21 Because Mas-deficient miceresent a marked change in the ECM protein expressiono a pro-fibrotic pattern,3 these findings indicate thatng-(1–7) acts directly via Mas present in cardiac fibro-lasts to exert its antifibrotic effects. Our present data aren accordance with these previous studies, because theardiac ECM proteins in TG rats, which are normoten-ive,15 were finely regulated by Ang-(1–7). The directffect of Ang-(1–7) on the fibroblast functions12 is cor-oborated by our findings, after a reduction of some ECMroteins was also observed in untreated TG rats that didot show any cardiac deleterious consequences caused bysoproterenol treatment. It is important to note that, de-endent of the experimental condition (treated or not withsoproterenol), Ang-(1–7) selectively regulated the me-

abolism of each ECM protein analyzed. A similar find- r

ng was also observed in previous studies.3,21 It remainso be established if Ang-(1–7) influences the synthesis oregradation of ECM proteins or both. In this aspect, theffects of Ang-(1–7) on the matrix metalloproteinasesetabolism should be considered in future studies. In

act, Pan et al22 have found that Ang-(1–7) and Ang IIave opposite effects on the regulation of matrix metal-oproteinases and tissue inhibitors of matrix metallopro-einases transcription in primary cultures of fibroblastsnd cardiomyocytes.22

In addition to its antifibrotic effect, Ang-(1–7) has beenlso described as an antitrophic peptide in the heart.7,10,12

allant et al10 have reported that Ang-(1–7) directly inhibitshe growth of cultured cardiomyocytes through activation of

as. Moreover, it has been demonstrated that the hypertro-hic effects of conditioned medium from Ang II–treateddult rat cardiac fibroblasts are attenuated in cardiomyo-ytes pretreated with Ang-(1–7).12 The antitrophic effect ofng-(1–7) was also viewed in our model, because in arevious study it was observed that the cardiac hypertrophicffect of isoproterenol was attenuated in TGR(A1–7)3292

Figure 3. Immunofluorescent localization of collagen typeIII in LVs. Low-magnification images reveal the overall levelof immunostaining in (A) SD vehicle, (B) SD ISO, (C)TGR(A1–7)3292 (TG) vehicle, and (D) TG ISO groups. (E)Negative control was obtained when the primary antibodywas omitted from the incubation procedure. Blue stainingcorresponds to nuclei labeled with 4=6-DAPI. Original mag-nification, 40�. (F) Quantification of collagen type III inLVs. Data are shown as means � SEM. A.U., arbitrary unit;DAPI, diamidino-2-phenylindole dihydrochloride; ISO, iso-proterenol; LV, left ventricle; SD, Sprague-Dawley. ***P �.001 (one-way analysis of variance followed by Newman-Keuls posttest).

ats.15 These data suggest that, besides its role in regulating

tis

EtAledirciSalosarRbpiTfil

nApnTS

t

eBprfgwLBcTsAAasaAldwolo

C

i

TLDi

L

STP

ST

r

TEaT

A

STA

STM

ST

S

a

rA

346 A.P. Nadu et al. / Journal of the American Society of Hypertension 2(5) (2008) 341–348

he ECM protein balance in the heart, Ang-(1–7) is alsomportantly involved in the maintenance of the myocytestructure.

It is well-established that Ang II increases the cardiacCM proteins content acting through AT1 receptors (1). On

he other hand, it has been reported that chronic infusion ofng-(1–7) in Wistar rats significantly decreases the Ang II

evels in the heart.23 In the present study, the antifibroticffect observed in TG rats was accompanied by a significantecrease in LV Ang II levels at basal conditions and aftersoproterenol treatment. Therefore, Ang-(1–7) could alsoegulate the ECM proteins content by decreasing the Ang IIoncentration in the heart. Also, isoproterenol treatmentnduced an increase in Ang II concentration in hearts fromD rats, but not in TG rats, further suggesting that thentifibrotic effect observed in TG rats involves a downregu-ation of cardiac Ang II. The regulatory action of Ang-(1–7)n the Ang II levels is apparently heart-specific, because noignificant alterations in the Ang II levels of other organs suchs lung, kidney, adrenal glands, and testis were observed in TGats (Ferreira AJ, Nadu AP, Reudelhuber TL, Bader M, SantosAS, unpublished data). A similar finding was also reportedy Mendes et al.23 Alternatively, Iwata et al12 have shown thatretreatment of adult rat cardiac fibroblasts with Ang-(1–7)nhibits the Ang II–induced increases in collagen synthesis.his possibility as well as a direct effect of Ang-(1–7) inbroblasts is particularly likely, considering that increases of

ocal Ang II did not change heart remodeling.24

The higher plasma Ang II levels observed in TG rats wasot fully investigated in the current study; however, lungCE expression and activity, plasma ACE activity, andlasma renin activity are not the cause of this effect, sinceo significant differences were observed between SD andG rats (Ferreira AJ, Nadu AP, Reudelhuber TL, Bader M,antos RAS, unpublished data).To identify possible mechanisms underlying the regula-

ion of the Ang II levels in the heart, we examined the

able 1eft ventricle and plasma angiotensin II levels in Sprague-awley and TGR(A1–7)3292 rats treated or not with

soproterenol

eft ventricle Angiotensin II (pg/mg protein)Vehicle ISO

D 1.06 � 0.07 (n � 6) 1.23 � 0.06* (n � 7)G 0.77 � 0.04†† (n � 6) 0.83 � 0.10†† (n � 7)lasma Angiotensin II (pg/mL)

Vehicle ISOD 76.69 � 10.14 (n � 8) 72.94 � 9.40 (n � 7)G 112.0 � 12.37 (n � 8)† 118.0 � 14.06 (n � 9)†

ISO, isoproterenol; SD, Sprague-Dawley; TG, TGR(A1–7)3292.The data are expressed as mean � SEM.* P � .05 when compared with vehicle-treated SD animals and† P � .05;†† P � .01 when compared with the corresponding SD control

Mats (Student’s t test).

xpression of some RAS components in SD and TG rats.ecause angiotensinogen is the precursor of the angiotensineptides, changes in its expression could explain the down-egulation of Ang II in hearts of TGR(A1–7)3292 rats. Inact, TG rats presented a lower expression of angiotensino-en mRNA in both basal and isoproterenol conditions. Also,e found that the expression of ACE2 is augmented in theV of TG rats compared with SD rats at basal conditions.ecause ACE2 is an Ang II–degrading enzyme, it couldontribute to the lower Ang II concentration in the hearts ofG rats. However, no significant changes in ACE2 expres-ion was observed in TG rats treated with isoproterenol.dditionally, isoproterenol treatment induced an increase ofCE2 expression only in SD rats. These findings are in

greement with previous reports showing that ACE2 iselectively regulated according to the pathologic stimulusnd to the duration of the injury.25,26 Furthermore, becauseCE2 is the main Ang-(1–7)–forming enzyme, an increased

ocal production of this peptide could be the cause of theownregulation of its receptor observed in SD rats treatedith isoproterenol. Nevertheless, changes in the expressionf Mas are not involved in the regulation of the Ang IIevels in TG rats, after its expression was similar to thatbserved in SD rats.

onclusion

A growing body of evidence suggests that the heart is anmportant target for the actions of the ACE2-Ang-(1–7)-

able 2ffects of isoproterenol treatment on angiotensinogen, ACE2nd Mas expression in left ventricles of Sprague-Dawley andGR(A1–7)3292 rats

CE2 expressionVehicle ISO

D 0.068 � 0.006 (n � 4) 0.142 � 0.013** (n � 4)G 0.151 � 0.033 (n � 4)† 0.159 � 0.035 (n � 4)ngiotensinogen expression

Vehicle ISOD 0.046 � 0.003 (n � 3) 0.045 � 0.001 (n � 3)G 0.031 � 0.001†† (n � 3) 0.032 � 0.005† (n � 3)as receptor expression

Vehicle ISOD 0.541 � 0.140 (n � 4) 0.140 � 0.067* (n � 4)G 0.483 � 0.140 (n � 5) 0.479 � 0.161 (n � 5)

ACE2, angiotensin-converting enzyme 2; ISO, isoproterenol;D, Sprague-Dawley; TG, TGR(A1–7)3292.The data are expressed as mean � SEM.* P � .05, ** P � .01 when compared with vehicle-treated SD

nimals and† P � .05,†† P � .01 when compared with the corresponding SD control

ats (Student’s t test). The results are reported as the ratio betweenCE2, angiotensinogen, or Mas and GAPDH values.

as axis. Among its cardiac effects, the antifibrotic/antitro-

priMsdeMts

R

1

1

1

1

1

1

1

1

1

1

2

2

347A.P. Nadu et al. / Journal of the American Society of Hypertension 2(5) (2008) 341–348

hic action is increasingly considered to play an importantole in the homeostasis of the heart. ACE2 overexpressionn the heart (20), Ang-(1–7) infusion,6,14 and deletion of

as receptor3 have validated this hypothesis. In the presenttudy, using a transgenic model, we provided further evi-ence for a key role of Ang-(1–7) in the modulation of thexpression of ECM proteins in the heart. Further studies inas�/� and ACE2�/� animals would be important to fur-

her pursuing the role of Ang-(1–7)/Mas/ACE2 in hearttructure and function.

eferences

1. Lijnen PJ, Petrov VV. Role of intracardiac renin-angio-tensin-aldosterone system in extracellular matrix re-modeling. Methods Find Exp Clin Pharmacol 2003;25:541–64.

2. Weber KT, Brilla CG. Pathological hypertrophy andcardiac interstitium. Fibrosis and renin-angiotensin-al-dosterone system. Circulation 1991;83:1849–65.

3. Santos RAS, Castro CH, Gava E, Pinheiro SV, AlmeidaAP, Paula RD, et al. Impairment of in vitro and in vivoheart function in angiotensin-(1–7) receptor Masknockout mice. Hypertension 2006;47:996–1002.

4. 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 cardio-vascular drugs. Expert Opin Investig Drugs 2005;14:1019–31.

5. Ferreira AJ, Santos RAS, Almeida AP. Angiotensin-(1–7): cardioprotective effect in myocardial ischemia/reperfusion. Hypertension 2001;38:665–68.

6. Grobe JL, Mecca AP, Lingis M, Shenoy V, Bolton TA,Machado JM, et al. Prevention of angiotensin II-in-duced cardiac remodeling by angiotensin-(1–7). Am JPhysiol Heart Circ Physiol 2007;292:H736–42.

7. Loot AE, Roks AJM, Henning RH, Tio RA, SuurmeijerAJH, Boomsma F, et al. Angiotensin-(1–7) attenuatesthe development of heart failure after myocardial in-farction in rats. Circulation 2002;105:1548–50.

8. Ferreira AJ, Jacoby BA, Araujo CA, Macedo FA, SilvaGAB, Almeida AP, et al. The nonpeptide angiotensin-(1–7) receptor Mas agonist AVE 0991 attenuates heartfailure induced by myocardial infarction. Am J PhysiolHeart Circ Physiol 2007;292:H1113–19.

9. Ferreira AJ, Santos RAS, Almeida AP. Angiotensin-(1–7) improves the post-ischemic function in isolatedperfused rat hearts. Braz J Med Biol Res 2002;35:1083–90.

0. Tallant EA, Ferrario CM, Gallagher PE. Angiotensin-(1–7) inhibits growth of cardiac myocytes through ac-tivation of the Mas receptor. Am J Physiol Heart Circ

Physiol 2005;289:H1560–66.

1. Santos RAS, Simões e Silva AC, Maric C, Silva DM,Machado RP, de Buhr I, et al. Angiotensin-(1–7) isan endogenous ligand for the G protein-coupled re-ceptor Mas. Proc Natl Acad Sci U S A 2003;100:8258 – 63.

2. Iwata M, Cowling RT, Gurantz D, Moore C, Zhang S,Yuan JX, et al. Angiotensin-(1–7) binds to specificreceptors on cardiac fibroblasts to initiate anti-fibroticand anti-trophic effects. Am J Physiol Heart CircPhysiol 2005;289:H2356–63.

3. Castro CH, Santos RAS, Ferreira AJ, Bader M, AleninaN, Almeida AP. Effects of genetic deletion of angio-tensin-(1–7) receptor Mas on cardiac function duringischemia/reperfusion in the isolated perfused mouseheart. Life Sci 2006;80:264–68.

4. Grobe JL, Mecca AP, Mao H, Katovich MJ. Chronicangiotensin-(1–7) prevents cardiac fibrosis in DOCA-salt model of hypertension. Am J Physiol Heart CircPhysiol 2006;290:H2417–23.

5. Santos RAS, Ferreira AJ, Nadu AP, Braga ANG,Almeida AP, Campagnole-Santos MJ, et al. Expressionof an angiotensin-(1–7)-producing fusion protein pro-duces cardioprotective effects in rats. Physiol Genom-ics 2004;17:292–99.

6. Botelho-Santos GA, Sampaio WO, Reudelhuber TL,Bader M, Campagnole-Santos MJ, Santos RAS. Ex-pression of an angiotensin-(1–7)-producing fusion pro-tein in rats induced marked changes in regional vascularresistance. Am J Physiol Heart Circ Physiol 2007;292:H2485–90.

7. Botelho LMO, Block CH, Khosla MC, Santos RAS.Plasma angiotensin-(1–7) immunoreactivity is in-creased by salt load, water deprivation, and hemor-rhage. Peptides 1994;15:723–29.

8. Bradford MM. A rapid and sensitive method for thequantification of microgram quantities of protein utiliz-ing the principle of protein-dye binding. Anal Biochem1976;72:248–54.

9. Benter IF, Yousif MH, Anim JT, Cojocel C, Diz DI.Angiotensin-(1–7) prevents development of severe hy-pertension and end-organ damage in spontaneously hy-pertensive rats treated with L-NAME. Am J PhysiolHeart Circ Physiol 2006;290:H684–91.

0. Diez-Freire C, Vazquez J, Correa de Adjounian MF,Ferrari MF, Yuan L, Silver X, et al. ACE2 genetransfer attenuates hypertension-linked pathophysio-logical changes in the SHR. Physiol Genomics 2006;27:12–9.

1. Ferreira AJ, Oliveira TL, Castro MC, Almeida AP,Castro CH, Caliari MV, et al. Isoproterenol-inducedimpairment of heart function and remodeling are atten-uated by the nonpeptide angiotensin-(1–7) analogue

AVE 0991. Life Sci 2007;81:916–23.

2

2

2

2

2

348 A.P. Nadu et al. / Journal of the American Society of Hypertension 2(5) (2008) 341–348

2. Pan CH, Wen CH, Lin CS. Interplay of angiotensin IIand angiotensin-(1–7) in the regulations of matrix met-alloproteinases of human cardiocytes. Exp Physiol2008;93:599–612.

3. Mendes AC, Ferreira AJ, Pinheiro SV, Santos RAS.Chronic infusion of angiotensin-(1–7) reduces heart angio-tensin II levels in rats. Regul Pept 2005;125:29–34.

4. Reudelhuber TL, Bernstein KE, Delafontaine P. Is angio-tensin II a direct mediator of left ventricular hypertrophy?

5. Ishiyama Y, Gallagher PE, Averill DB, Tallant EA,Brosnihan KB, Ferrario CM. Upregulation of angioten-sin-converting enzyme 2 after myocardial infarction byblockade of angiotensin II receptors. Hypertension2004;43:970–76.

6. Ocaranza MP, Godoy I, Jalil JE, Varas M, Collantes P,Pinto M, et al. Enalapril attenuates downregulation ofangiotensin-converting enzyme 2 in the late phase ofventricular dysfunction in myocardial infarcted rat. Hy-

Time for another look. Hypertension 2007;49:1196–201. pertension 2006;48:572–78.

Receive Tables of Contents by e-mail

To receive Tables of Contents by e-mail, sign up through our website at www.ashjournal.com

InstructionsLog on to www.ashjournal.com and click on the “Add Table of Contents Alert” link in the Receive Free E-mail Alertssection of the home page. Complete the registration process. You will receive an e-mail message confirming that youhave been added to the mailing list. Note that the Table of Contents e-mails will be sent when a new issue is postedto the Web.