Network


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

Hotspot


Dive into the research topics where Felipe C.G. Reis is active.

Publication


Featured researches published by Felipe C.G. Reis.


Biological Chemistry | 2007

Increase in kinins on post-exercise hypotension in normotensive and hypertensive volunteers.

Milton Rocha Moraes; Reury Frank Pereira Bacurau; João D.S. Ramalho; Felipe C.G. Reis; Dulce Elena Casarini; Jair R. Chagas; Vitor Oliveira; Elisa Mieko Suemitsu Higa; Dulcineia S.P. Abdalla; Jorge L. Pesquero; João Bosco Pesquero; Ronaldo C. Araujo

Abstract Post-exercise hypotension is an important event for blood pressure regulation, especially in hypertensive individuals. Although post-exercise hypotension is a well-known phenomenon, the mechanism responsible is still unclear. The kallikrein-kinin system is involved in blood pressure control, but its role in post-exercise hypotension has not yet been investigated. Thus, the purpose of this study was to investigate the involvement of the vasodilators bradykinin and des-Arg9-BK and kallikrein activity in post-exercise hypotension promoted by 35 min of cycle ergometer (CE) or circuit weight-training (CWT) bouts in normotensive and hypertensive individuals. A significant decrease in mean arterial pressure at 45 and 60 min after CE and 45 min after CWT was observed in normotensive individuals. Hypertensive values of mean arterial pressure were significantly reduced at 45 and 60 min after CE and at 60 min after CWT. Before exercise, plasma bradykinin concentrations and kallikrein activity were higher in hypertensive compared to normotensive volunteers. Kinin levels increased in the groups evaluated at the end of the training period and 60 min post-exercise. These data suggest that the kallikrein-kinin system may be involved in post-exercise hypotension in normotensive and hypertensive individuals subjected to CE and CWT bouts.


International Immunopharmacology | 2008

Effect of angiotensin converting enzyme inhibitor enalapril on body weight and composition in young rats

Edson Lucas dos Santos; Kely de Picoli Souza; Paola Bianchi Guimarães; Felipe C.G. Reis; Sylvia Maria Affonso da Silva; Claudio M. Costa-Neto; Jacqueline Luz; João Bosco Pesquero

Obesity is considered a worldwide public health problem showing an increased prevalence in developing countries, with urgent need for new and more efficient drugs and therapies. Enalapril, an angiotensin-I converting enzyme inhibitor (ACEi), is classically used in anti-hypertensive therapies, however, earlier publications have shown that this drug could also have significant impact on body weight in rats as well as in humans, besides reducing blood pressure. The effect of this drug in the white adipose tissue has been neglected for long time, even considering that most components of the renin-angiotensin and kallikrein-kinin system are expressed in this tissue. Furthermore, the adipose tissue is considered today as one of the most important sites for endocrine/inflammatory regulation of appetite and energy output and AngII has been linked to the metabolism in this tissue. Therefore, we analyzed the influence of chronic enalapril treatment in normotensive rats at earlier ages, evaluating body weight, energy homeostasis, lipid profile and serum levels of the hormones leptin and insulin, in the presence of a standard or a palatable hyperlipidic diet regimen for one month. Our results show that enalapril treatment is able to reduce body fat on both diets, without alteration in serum lipid profile. Furthermore, animals receiving enalapril showed reduction in food intake, leptin level and energy intake. In summary, these findings show for the first time that the ACEi enalapril reduces body fat in young normotensive rats and highlights a novel target to treat obesity and associated diseases.


Diabetes | 2008

Kinin B1 Receptor Deficiency Leads to Leptin Hypersensitivity and Resistance to Obesity

Marcelo A. Mori; Ronaldo C. Araujo; Felipe C.G. Reis; Daniela G. Sgai; Raphael Gomes Fonseca; Carlos C. Barros; Vanessa F. Merino; Mariana Passadore; Ana M.R.B. Barbosa; Bernard Ferrari; Pierre Carayon; Charlles Heldan de Moura Castro; Suma I. Shimuta; Jacqueline Luz; Jean-Loup Bascands; Joost P. Schanstra; Patrick Even; Suzana M. Oliveira; Michael Bader; João Bosco Pesquero

OBJECTIVE—Kinins mediate pathophysiological processes related to hypertension, pain, and inflammation through the activation of two G-protein–coupled receptors, named B1 and B2. Although these peptides have been related to glucose homeostasis, their effects on energy balance are still unknown. RESEARCH DESIGN AND METHODS—Using genetic and pharmacological strategies to abrogate the kinin B1 receptor in different animal models of obesity, here we present evidence of a novel role for kinins in the regulation of satiety and adiposity. RESULTS—Kinin B1 receptor deficiency in mice (B1−/−) resulted in less fat content, hypoleptinemia, increased leptin sensitivity, and robust protection against high-fat diet–induced weight gain. Under high-fat diet, B1−/− also exhibited reduced food intake, improved lipid oxidation, and increased energy expenditure. Surprisingly, B1 receptor deficiency was not able to decrease food intake and adiposity in obese mice lacking leptin (ob/ob-B1−/−). However, ob/ob-B1−/− mice were more responsive to the effects of exogenous leptin on body weight and food intake, suggesting that B1 receptors may be dependent on leptin to display their metabolic roles. Finally, inhibition of weight gain and food intake by B1 receptor ablation was pharmacologically confirmed by long-term administration of the kinin B1 receptor antagonist SSR240612 to mice under high-fat diet. CONCLUSIONS—Our data suggest that kinin B1 receptors participate in the regulation of the energy balance via a mechanism that could involve the modulation of leptin sensitivity.


Hypertension | 2008

ACE Activity Is Modulated by Kinin B2 Receptor

Regiane A. Sabatini; Paola Bianchi Guimarães; Liliam Fernandes; Felipe C.G. Reis; Patrícia Alessandra Bersanetti; Marcelo A. Mori; Alberto Navarro; Aline M. Hilzendeger; Edson Lucas dos Santos; Maria Claudina Camargo de Andrade; Jair R. Chagas; Jorge L. Pesquero; Dulce Elena Casarini; Michael Bader; Adriana K. Carmona; João Bosco Pesquero

Angiotensin-converting enzyme (ACE) is an ectoprotein able to modulate the activity of a plethora of compounds, among them angiotensin I and bradykinin. Despite several decades of research, new aspects of the mechanism of action of ACE have been elucidated, expanding our understanding of its role not only in cardiovascular regulation but also in different areas. Recent findings have ascribed an important role for ACE/kinin B2 receptor heterodimerization in the pharmacological properties of the receptor. In this work, we tested the hypothesis that this interaction also affects ACE enzymatic activity. ACE catalytic activity was analyzed in Chinese hamster ovary cell monolayers coexpressing the somatic form of the enzyme and the receptor coding region using as substrate the fluorescence resonance energy transfer peptide Abz-FRK(Dnp)P-OH. Results show that the coexpression of the kinin B2 receptor leads to an augmentation in ACE activity. In addition, this effect could be blocked by the B2 receptor antagonist icatibant. The hypothesis was also tested in endothelial cells, a more physiological system, where both proteins are naturally expressed. Endothelial cells from genetically ablated kinin B2 receptor mice showed a decreased ACE activity when compared with wild-type mice cells. In summary, this is the first report showing that the ACE/kinin B2 receptor interaction modulates ACE activity. Taking into account the interplay among ACE, ACE inhibitors, and kinin receptors, we believe that these results will shed new light into the arena of the controversial search for the mechanism controlling these interactions.


Peptides | 2011

Role of vascular Kinin B1 and B2 receptors in endothelial nitric oxide metabolism

Rodrigo A. Loiola; Felipe C.G. Reis; Elisa Mitiko Kawamoto; Cristoforo Scavone; Dulcineia S.P. Abdalla; Liliam Fernandes; João Bosco Pesquero

Kinin B(1) and B(2) receptors play an essential role in inflammatory process and cardiovascular homeostasis. The present study investigated the vascular reactivity and nitric oxide (NO) generation in the isolated mesenteric arteriolar bed from B(1) (B(1)(-/-)) and B(2) receptor (B(2)(-/-)) knockout mice. Endothelial-dependent relaxation was significantly decreased in arterioles from both B(1)(-/-) and B(2)(-/-) in comparison to wild type (WT) mice, with no differences for endothelial-independent relaxating or vasoconstrictor agents. Plasmatic and vascular NO production were markedly reduced in both B(1)(-/-) and B(2)(-/-). In contrast, in the presence of l-arginine, Ca(2+) and co-factors for the enzyme, NO synthase activity was higher in homogenates of mesenteric vessels of B(1)(-/-) and B(2)(-/-). The present study demonstrated that targeted deletion of B(1) or B(2) receptor gene in mice induces important alterations in the vascular reactivity of resistance vessels and NO metabolism. The severe impairment in the endothelial-mediated vasodilation accompanied by decreased NO bioavailability, despite the augmented NOS activity, strongly indicates an exacerbation of NO inactivation in B(1)(-/-) and B(2)(-/-) vessels. The present data provide valuable information in order to clarify the relevance of kinin receptors in regulating vascular physiology and may point to new approaches regarding its correlation with endothelial dysfunction, oxidative stress and NO availability.


Laboratory Investigation | 2012

Bradykinin inhibits hepatic gluconeogenesis in obese mice

Carlos C. Barros; Anderson Sola Haro; F.J. Russo; Ines Schadock; Sandro Soares de Almeida; Felipe C.G. Reis; Milton Rocha Moraes; Andre Abou Haidar; Aparecida Emiko Hirata; Marcelo A. Mori; Reury Frank Pereira Bacurau; Martin Würtele; Michael Bader; João Bosco Pesquero; Ronaldo C. Araujo

The kallikrein–kinin system (KKS) has been previously linked to glucose homeostasis. In isolated muscle or fat cells, acute bradykinin (BK) stimulation was shown to improve insulin action and increase glucose uptake by promoting glucose transporter 4 translocation to plasma membrane. However, the role for BK in the pathophysiology of obesity and type 2 diabetes remains largely unknown. To address this, we generated genetically obese mice (ob/ob) lacking the BK B2 receptor (obB2KO). Despite similar body weight or fat accumulation, obB2KO mice showed increased fasting glycemia (162.3±28.2 mg/dl vs 85.3±13.3 mg/dl), hyperinsulinemia (7.71±1.75 ng/ml vs 4.09±0.51 ng/ml) and impaired glucose tolerance when compared with ob/ob control mice (obWT), indicating insulin resistance and impaired glucose homeostasis. This was corroborated by increased glucose production in response to a pyruvate challenge. Increased gluconeogenesis was accompanied by increased hepatic mRNA expression of forkhead box protein O1 (FoxO1, four-fold), peroxisome proliferator-activated receptor gamma co-activator 1-alpha (seven-fold), phosphoenolpyruvate carboxykinase (PEPCK, three-fold) and glucose-6-phosphatase (eight-fold). FoxO1 nuclear exclusion was also impaired, as the obB2KO mice showed increased levels of this transcription factor in the nucleus fraction of liver homogenates during random feeding. Intraportal injection of BK in lean mice was able to decrease the hepatic mRNA expression of FoxO1 and PEPCK. In conclusion, BK modulates glucose homeostasis by affecting hepatic glucose production in obWT. These results point to a protective role of the KKS in the pathophysiology of type 2 diabetes mellitus.


Aging-us | 2016

Fat-specific Dicer deficiency accelerates aging and mitigates several effects of dietary restriction in mice

Felipe C.G. Reis; Jéssica L. O. Branquinho; Bruna B. Brandão; Beatriz A. Guerra; Ismael D. Silva; Andrea Frontini; Thomas Thomou; Loris Sartini; Saverio Cinti; C. Ronald Kahn; William T. Festuccia; Alicia J. Kowaltowski; Marcelo A. Mori

Aging increases the risk of type 2 diabetes, and this can be prevented by dietary restriction (DR). We have previously shown that DR inhibits the downregulation of miRNAs and their processing enzymes - mainly Dicer - that occurs with aging in mouse white adipose tissue (WAT). Here we used fat-specific Dicer knockout mice (AdicerKO) to understand the contributions of adipose tissue Dicer to the metabolic effects of aging and DR. Metabolomic data uncovered a clear distinction between the serum metabolite profiles of Lox control and AdicerKO mice, with a notable elevation of branched-chain amino acids (BCAA) in AdicerKO. These profiles were associated with reduced oxidative metabolism and increased lactate in WAT of AdicerKO mice and were accompanied by structural and functional changes in mitochondria, particularly under DR. AdicerKO mice displayed increased mTORC1 activation in WAT and skeletal muscle, where Dicer expression is not affected. This was accompanied by accelerated age-associated insulin resistance and premature mortality. Moreover, DR-induced insulin sensitivity was abrogated in AdicerKO mice. This was reverted by rapamycin injection, demonstrating that insulin resistance in AdicerKO mice is caused by mTORC1 hyperactivation. Our study evidences a DR-modulated role for WAT Dicer in controlling metabolism and insulin resistance.


Neuropeptides | 2010

Effect of kinin B2 receptor ablation on skeletal muscle development and myostatin gene expression

K. de Picoli Souza; Elice C. Batista; Elton Dias da Silva; Felipe C.G. Reis; Sylvia Maria Affonso da Silva; Ronaldo C. Araujo; Jônio Arruda Luz; Edson Lucas dos Santos; João Bosco Pesquero

Bradykinin (BK) is an active peptide that binds to the kinin B(2) receptor and induces biological events during the development and adult life. In this study we aimed to investigate the effect of kinin B(2) receptor ablation in the postnatal skeletal muscle development and body composition in adult life. For studies of skeletal muscle development, control (C57Bl6 - WT) and B(2) receptor knockout mice (B(2)(-/-)) were sacrificed at 15, 30 and 90days after birth, the gastrocnemius skeletal muscle was weighed and myostatin gene expression evaluated by real time PCR. For energy balance determination, data from control and B(2)(-/-) at 90 and 120days were collected by calorimetric method. Body composition at 120days was determined by chloroform-methanol (total body fat) and Lowry-modified method (total body protein). The results show that B(2)(-/-) have significantly increased total body weight at 15, 30 and 90days of life, when compared to WT. The weight of the gastrocnemius skeletal muscle was also significantly increased at 30 and 90days of life. Body composition analyses revealed that B(2)(-/-) mice exhibit more total corporal protein and less total corporal fat. Energy balance revealed that B(2)(-/-) have increased metabolizable energy intake and energy expenditure when compared to control mice, resulting in a lower energy gain. Interestingly, myostatin mRNA expression was significantly decreased in 15 and 30days old B(2)(-/-) mice and after icatibant treatment of WT adult mice for 5days. In conclusion, together our results show that kinin B(2) receptor deletion increases lean mass, reduces fat mass and improves metabolism efficiency in mice. The mechanism involved in this phenotype could be related to the reduction of myostatin gene expression during postnatal life.


Chemico-Biological Interactions | 2010

Myelopoiesis modulation by ACE hyperfunction in kinin B1 receptor knockout mice: Relationship with AcSDKP levels

Carlos Rocha Oliveira; Edgar J. Paredes-Gamero; Christiano M.V. Barbosa; Fábio D. Nascimento; Elice C. Batista; Felipe C.G. Reis; Antonio H. Martins; Alice T. Ferreira; Adriana K. Carmona; João Bosco Pesquero; Ivarne L.S. Tersariol; Ronaldo C. Araujo; Claudia Bincoletto

Angiotensin I-converting enzyme (ACE), a common element of renin-angiotensin system (RAS) and kallikrein-kinin system (KKS), is involved in myelopoiesis modulation, mainly by cleaving the tetrapeptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP). Based on this finding and in our results showing B1 and B2 kinin receptors expression in murine bone marrow (BM) cells, we evaluated the ACE influence on myelopoiesis of kinin B1 receptor knockout mice (B1KO) using long-term bone marrow cultures (LTBMCs). Captopril and AcSDKP were used as controls. Enhanced ACE activity, expressed by non-hematopoietic cells (Ter-199(-) and CD45(-)), was observed in B1KO LTBMCs when compared to wild-type (WT) cells. ACE hyperfunction in B1KO cells was maintained when LTBMCs from B1KO mice were treated with captopril (1.0microM) or AcSDKP (1.0nM). Although no alterations were observed in ACE mRNA and protein levels under these culture conditions, 3.0nM of AcSDKP increased ACE mRNA levels in WT LTBMCs. No alteration in the number of GM-CFC was seen in B1KO mice compared to WT animals, even when the former were treated with AcSDKP (10microg/kg) or captopril (100mg/kg) for 4 consecutive days. Hematological data also revealed no differences between WT and B1KO mice under basal conditions. When the animals received 4 doses of lipopolysaccharide (LPS), a decreased number of blood cells was detected in B1KO mice in relation to WT. We also found a decreased percentage of Gr1(+)/Mac-1(+), Ter119(+), B220(+), CD3(+), and Lin(-)Sca1(+)c-Kit(+) (LSK) cells in the BM of B1KO mice compared to WT animals. Low AcSDKP levels were observed in BM cultures from B1KO in comparison to WT cultures. We conclude that ACE hyperfunction in B1KO mice resulted in faster hydrolysis of AcSDKP peptide, which in turn decreased in BM tissues allowing HSC to enter the S stage of the cell cycle.


International Immunopharmacology | 2008

Swimming training exacerbates pathological cardiac hypertrophy in kinin B2 receptor-deficient mice

Elice C. Batista; Eliza C. Batista; João D.S. Ramalho; Felipe C.G. Reis; Carlos C. Barros; Milton Rocha Moraes; Jorge L. Pesquero; Reury Fp Bacurau; João Bosco Pesquero; Ronaldo C. Araujo

Kallikrein-kinin system exerts cardioprotective effects against pathological hypertrophy. These effects are modulated mainly via B2 receptor activation. Chronic physical exercise can induce physiological cardiac hypertrophy characterized by normal organization of cardiac structure. Therefore, the aim of this work was to verify the influence of kinin B2 receptor deletion on physiological hypertrophy to exercise stimulus. Animals were submitted to swimming practice for 5 min or for 60 min, 5 days a week, during 1 month and several cardiac parameters were evaluated. Results showed no significantly difference in heart weight between both groups, however an increased left ventricle weight and myocyte diameter were observed after the 60 min swimming protocol, which was more pronounced in B2(-/-) mice. In addition, sedentary B2(-/-) animals presented higher left ventricle mass when compared to wild-type (WT) mice. An increase in capillary density was observed in exercised animals, however the effect was less pronounced in B2(-/-) mice. Collagen, a marker of pathological hypertrophy, was increased in B2(-/-) mice submitted to swimming protocol, as well as left ventricular thickness, suggesting that these animals do not respond with physiological hypertrophy for this kind of exercise. In conclusion, our data suggest an important role for the kinin B2 receptor in physiological cardiac hypertrophy.

Collaboration


Dive into the Felipe C.G. Reis's collaboration.

Top Co-Authors

Avatar

João Bosco Pesquero

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Ronaldo C. Araujo

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Marcelo A. Mori

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Edson Lucas dos Santos

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Elice C. Batista

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Jorge L. Pesquero

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

Carlos C. Barros

Universidade Federal de Pelotas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liliam Fernandes

Federal University of São Paulo

View shared research outputs
Researchain Logo
Decentralizing Knowledge