Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Anderson J. Ferreira is active.

Publication


Featured researches published by Anderson J. Ferreira.


Experimental Physiology | 2008

Recent advances in the angiotensin‐converting enzyme 2–angiotensin(1–7)–Mas axis

Robson A.S. Santos; Anderson J. Ferreira; Ana Cristina Simões e Silva

In the past few years, the classical concept of the renin–angiotensin system (RAS) has experienced substantial conceptual changes. The identification of: the renin/prorenin receptor; the angiotensin‐converting enzyme homologue, ACE2, as an angiotensin peptide‐processing enzyme and a virus receptor for severe acute respiratory syndrome, the Mas as a receptor for angiotensin (1–7) [Ang(1–7)], and the possibility of signaling through ACE have contributed to switch our understanding of the RAS from the classical limited‐proteolysis linear cascade to a cascade with multiple mediators, multiple receptors and multifunctional enzymes. With regard to Ang(1–7), the identification of ACE2 and of Mas as a receptor implicated in its actions contributed to decisively establish this heptapeptide as a biologically active member of the RAS cascade. In this review, we will focus on the recent findings related to the ACE2–Ang(1–7)–Mas axis and, in particular, on its putative role as an ACE–Ang II–AT1 receptor counter‐regulatory axis within the RAS.


Journal of Endocrinology | 2013

Angiotensin-converting enzyme 2, angiotensin-(1–7) and Mas: new players of the renin–angiotensin system

Robson A.S. Santos; Anderson J. Ferreira; Thiago Verano-Braga; Michael Bader

Angiotensin (Ang)-(1-7) is now recognized as a biologically active component of the renin-angiotensin system (RAS). Ang-(1-7) appears to play a central role in the RAS because it exerts a vast array of actions, many of them opposite to those attributed to the main effector peptide of the RAS, Ang II. The discovery of the Ang-converting enzyme (ACE) homolog ACE2 brought to light an important metabolic pathway responsible for Ang-(1-7) synthesis. This enzyme can form Ang-(1-7) from Ang II or less efficiently through hydrolysis of Ang I to Ang-(1-9) with subsequent Ang-(1-7) formation by ACE. In addition, it is now well established that the G protein-coupled receptor Mas is a functional binding site for Ang-(1-7). Thus, the axis formed by ACE2/Ang-(1-7)/Mas appears to represent an endogenous counterregulatory pathway within the RAS, the actions of which are in opposition to the vasoconstrictor/proliferative arm of the RAS consisting of ACE, Ang II, and AT(1) receptor. In this brief review, we will discuss recent findings related to the biological role of the ACE2/Ang-(1-7)/Mas arm in the cardiovascular and renal systems, as well as in metabolism. In addition, we will highlight the potential interactions of Ang-(1-7) and Mas with AT(1) and AT(2) receptors.


Hypertension | 2001

Angiotensin-(1-7): Cardioprotective Effect in Myocardial Ischemia/Reperfusion

Anderson J. Ferreira; Robson A.S. Santos; Alvair P. Almeida

In this study we evaluate the effects of angiotensin-(1-7) on reperfusion arrhythmias in isolated rat hearts. Rat hearts were perfused according to Langendorff technique and maintained in heated (37±1°C) and continuously gassed (95% O2/5% CO2) Krebs-Ringer solution at constant pressure (65 mm Hg). The electrical activity was recorded with an ECG (bipolar). Local ischemia was induced by coronary ligation for 15 minutes. After ischemia, hearts were reperfused for 30 minutes. Cardiac arrhythmias were defined as the presence of ventricular tachycardia and/or ventricular fibrillation after the ligation of the coronary artery was released. Angiotensin II (0.20 nmol/L, n=10) produced a significant enhancement of reperfusion arrhythmias. On the other hand, Ang-(1-7) presented in the perfusion solution (0.22 nmol/L, n=11) reduced incidence and duration of arrhythmias. The antiarrhythmogenic effects of Ang-(1-7) was blocked by the selective Ang-(1-7) antagonist A-779 (2 nmol/L, n=9) and by indomethacin pretreatment (5 mg/kg IP, n=8) but not by the bradykinin B2 antagonist HOE 140 (100 nmol/L, n=10) or by L-NAME pretreatment (30 mg/kg IP, n=8). These results suggest that the antiarrhythmogenic effect of low concentrations of Ang-(1-7) is mediated by a specific receptor and that release of endogenous prostaglandins .by Ang-(1-7) contributes to the alleviation of reversible and/or irreversible ischemia-reperfusion injury.


Hypertension | 2006

Impairment of In Vitro and In Vivo Heart Function in Angiotensin-(1-7) Receptor Mas Knockout Mice

Robson A.S. Santos; Carlos H. Castro; Elisandra Gava; Sérgio V.B. Pinheiro; Alvair P. Almeida; Renata Dutra de Paula; Jader Santos Cruz; Anderson S. Ramos; Kaleizu Teodoro Rosa; M.C. Irigoyen; Michael Bader; Natalia Alenina; Gregory T. Kitten; Anderson J. Ferreira

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.


Circulation Research | 2008

Angiotensin(1-7) Blunts Hypertensive Cardiac Remodeling by a Direct Effect on the Heart

Chantal Mercure; Alvaro Yogi; Glaucia E. Callera; Anna B. Aranha; Michael Bader; Anderson J. Ferreira; Robson A.S. Santos; Thomas Walther; Rhian M. Touyz; Timothy L. Reudelhuber

Angiotensin-converting enzyme 2 (ACE2) converts the vasopressor angiotensin II (Ang II) into angiotensin (1-7) [Ang(1-7)], a peptide reported to have vasodilatory and cardioprotective properties. Inactivation of the ACE2 gene in mice has been reported by one group to result in an accumulation of Ang II in the heart and an age-related defect in cardiac contractility. A second study confirmed the role of ACE2 as an Ang II clearance enzyme but failed to reproduce the contractility defects previously reported in ACE2-deficient mice. The reasons for these differences are unclear but could include differences in the accumulation of Ang II or the deficiencies in Ang(1-7) in the mouse models used. As a result, the roles of ACE2, Ang II, and Ang(1-7) in the heart remain controversial. Using a novel strategy, we targeted the chronic overproduction of either Ang II or Ang(1-7) in the heart of transgenic mice and tested their effect on age-related contractility and on cardiac remodeling in response to a hypertensive challenge. We demonstrate that a chronic accumulation of Ang II in the heart does not result in cardiac contractility defects, even in older (8-month-old) mice. Likewise, transgenic animals with an 8-fold increase in Ang(1-7) peptide in the heart exhibited no differences in resting blood pressure or cardiac contractility as compared to age-matched controls, but they had significantly less ventricular hypertrophy and fibrosis than their nontransgenic littermates in response to a hypertensive challenge. Analysis of downstream signaling cascades demonstrates that cardiac Ang(1-7) selectively modulates some of the downstream signaling effectors of cardiac remodeling. These results suggest that Ang(1-7) can reduce hypertension-induced cardiac remodeling through a direct effect on the heart and raise the possibility that pathologies associated with ACE2 inactivation are mediated in part by a decrease in production of Ang(1-7).


American Journal of Respiratory and Critical Care Medicine | 2009

Evidence for angiotensin-converting enzyme 2 as a therapeutic target for the prevention of pulmonary hypertension.

Anderson J. Ferreira; Vinayak Shenoy; Yoriko Yamazato; Srinivas Sriramula; Joseph Francis; Lihui Yuan; Ronald K. Castellano; David A. Ostrov; Suk Paul Oh; Michael J. Katovich; Mohan K. Raizada

RATIONALE It has been proposed that an activated renin angiotensin system (RAS) causes an imbalance between the vasoconstrictive and vasodilator mechanisms involving the pulmonary circulation leading to the development of pulmonary hypertension (PH). Recent studies have indicated that angiotensin-converting enzyme 2 (ACE2), a member of the vasoprotective axis of the RAS, plays a regulatory role in lung pathophysiology, including pulmonary fibrosis and acute lung disease. Based on these observations, we propose the hypothesis that activation of endogenous ACE2 can shift the balance from the vasoconstrictive, proliferative axis (ACE-Ang II-AT1R) to the vasoprotective axis [ACE2-Ang-(1-7)-Mas] of the RAS, resulting in the prevention of PH. OBJECTIVES We have taken advantage of a recently discovered synthetic activator of ACE2, XNT (1-[(2-dimethylamino) ethylamino]-4-(hydroxymethyl)-7-[(4-methylphenyl) sulfonyl oxy]-9H-xanthene-9-one), to study its effects on monocrotaline-induced PH in rats to support this hypothesis. METHODS The cardiopulmonary effects of XNT were evaluated in monocrotaline-induced PH rat model. MEASUREMENTS AND MAIN RESULTS A single subcutaneous treatment of monocrotaline in rats resulted in elevated right ventricular systolic pressure, right ventricular hypertrophy, increased pulmonary vessel wall thickness, and interstitial fibrosis. These changes were associated with increases in the mRNA levels of renin, ACE, angiotensinogen, AT1 receptors, and proinflammatory cytokines. All these features of PH were prevented in these monocrotaline-treated rats by chronic treatment with XNT. In addition, XNT caused an increase in the antiinflammatory cytokine, IL-10. CONCLUSIONS These observations provide conceptual support that activation of ACE2 by a small molecule can be a therapeutically relevant approach for treating and controlling PH.


Hypertension | 2008

Structure-Based Identification of Small-Molecule Angiotensin-Converting Enzyme 2 Activators as Novel Antihypertensive Agents

Jose A. Hernandez Prada; Anderson J. Ferreira; Michael J. Katovich; Vinayak Shenoy; Yanfei Qi; Robson A.S. Santos; Ronald K. Castellano; Andrew J. Lampkins; Vladimir Gubala; David A. Ostrov; Mohan K. Raizada

Angiotensin-converting enzyme 2 (ACE2) is a key renin-angiotensin system enzyme involved in balancing the adverse effects of angiotensin II on the cardiovascular system, and its overexpression by gene transfer is beneficial in cardiovascular disease. Therefore, our objectives were 2-fold: to identify compounds that enhance ACE2 activity using a novel conformation-based rational drug discovery strategy and to evaluate whether such compounds reverse hypertension-induced pathophysiologies. We used a unique virtual screening approach. In vitro assays revealed 2 compounds (a xanthenone and resorcinolnaphthalein) that enhanced ACE2 activity in a dose-dependent manner. Acute in vivo administration of the xanthenone resulted in a dose-dependent transient and robust decrease in blood pressure (at 10 mg/kg, spontaneously hypertensive rats decreased 71±9 mm Hg and Wistar-Kyoto rats decreased 21±8 mm Hg; P<0.05). Chronic infusion of the xanthenone (120 μg/day) resulted in a modest decrease in the spontaneously hypertensive rat blood pressure (17 mm Hg; 2-way ANOVA; P<0.05), whereas it had no effect in Wistar-Kyoto rats. Strikingly, the decrease in blood pressure was also associated with improvements in cardiac function and reversal of myocardial, perivascular, and renal fibrosis in the spontaneously hypertensive rats. We conclude that structure-based screening can help identify compounds that activate ACE2, decrease blood pressure, and reverse tissue remodeling. Administration of ACE2 activators may be a valid strategy for antihypertensive therapy.


Hypertension | 2005

Evidence for a Functional Interaction of the Angiotensin-(1–7) Receptor Mas With AT1 and AT2 Receptors in the Mouse Heart

Carlos H. Castro; Robson Augusto Souza dos Santos; Anderson J. Ferreira; Michael Bader; Natalia Alenina; Alvair P. Almeida

The aim of this study was to evaluate the angiotensin (Ang)-(1–7) effects in isolated mouse hearts. The hearts of male C57BL/6J and knockout mice for the Ang-(1–7) receptor Mas were perfused by the Langendorff method. After a basal period, the hearts were perfused for 20 minutes with Krebs-Ringer solution (KRS) alone (control) or KRS containing Ang-(1–7) (0.22 pmol/L), the Mas antagonist A-779 (115 nmol/L), the angiotensin type 1 receptor antagonist losartan (2.2 &mgr;mol/L), or the angiotensin type 2 receptor antagonist PD123319 (130 nmol/L). To evaluate the involvement of Ang receptors, prostaglandins, and nitric oxide in the Ang-(1–7) effects, the hearts were perfused for 20 to 30 minutes with KRS containing either A-779, losartan, PD123319, indomethacin, or NG-nitro-l-arginine methyl ester (l-NAME) alone or in association with subsequent Ang-(1–7) perfusion. In addition, hearts from Mas-knockout mice were perfused for 20 minutes with KRS containing Ang-(1–7) (0.22 pmol/L) and losartan. Ang-(1–7) alone did not change the perfusion pressure. Strikingly, in the presence of losartan, 0.22 pmol/L Ang-(1–7) induced a significant decrease in perfusion pressure, which was blocked by A-779, indomethacin, and l-NAME. Furthermore, this effect was not observed in Mas-knockout mice. In contrast, in the presence of PD123319, Ang-(1–7) produced a significant increase in perfusion pressure. This change was not modified by the addition of A-779. Losartan reduced but did not abolish this effect. Our results suggest that Ang-(1–7) produces complex vascular effects in isolated, perfused mouse hearts involving interaction of its receptor with angiotensin type 1- and type 2-related mechanisms, leading to the release of prostaglandins and nitric oxide.


Brazilian Journal of Medical and Biological Research | 2005

Cardiovascular actions of angiotensin-(1-7)

Anderson J. Ferreira; Robson A.S. Santos

Angiotensin-(1-7) (Ang-(1-7)) is now considered to be a biologically active member of the renin-angiotensin system. The functions of Ang-(1-7) are often opposite to those attributed to the main effector component of the renin-angiotensin system, Ang II. Chronic administration of angiotensin-converting enzyme inhibitors (ACEI) increases 10- to 25-fold the plasma levels of this peptide, suggesting that part of the beneficial effects of ACEI could be mediated by Ang-(1-7). Ang-(1-7) can be formed from Ang II or directly from Ang I. Other enzymatic pathways for Ang-(1-7) generation have been recently described involving the novel ACE homologue ACE2. This enzyme can form Ang-(1-7) from Ang II or less efficiently by the hydrolysis of Ang I to Ang-(1-9) with subsequent Ang-(1-7) formation. The biological relevance of Ang-(1-7) has been recently reinforced by the identification of its receptor, the G-protein-coupled receptor Mas. Heart and blood vessels are important targets for the formation and actions of Ang-(1-7). In this review we will discuss recent findings concerning the biological role of Ang-(1-7) in the heart and blood vessels, taking into account aspects related to its formation and effects on these tissues. In addition, we will discuss the potential of Ang-(1-7) and its receptor as a target for the development of new cardiovascular drugs.


Current Opinion in Nephrology and Hypertension | 2007

Angiotensin-(1-7) and the renin-angiotensin system.

Robson As Santos; Anderson J. Ferreira

Purpose of reviewIn this review we will focus on the recent findings related to angiotensin-(1–7) as an angiotensin II counter-regulatory peptide within the renin–angiotensin system. Recent findingsThe identification of the angiotensin-converting enzyme homologue ACE2 as an angiotensin peptide processing enzyme and of Mas as a receptor for angiotensin-(1–7) has contributed to establishing this heptapeptide as a biologically active member of the renin–angiotensin system cascade. SummaryThe previously unsuspected complexity of the renin–angiotensin system, unmasked by novel findings, has revealed new possibilities for exploring its physiological and pathophysiological roles. In addition, the ACE2–angiotensin-(1–7)–Mas axis may be seriously considered as a putative target for the development of new cardiovascular drugs.

Collaboration


Dive into the Anderson J. Ferreira's collaboration.

Top Co-Authors

Avatar

Robson A.S. Santos

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alvair P. Almeida

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Michael Bader

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlos H. Castro

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Giselle Foureaux

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Marcelo Vidigal Caliari

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Rubén D. Sinisterra

Universidade Federal de Minas Gerais

View shared research outputs
Researchain Logo
Decentralizing Knowledge