Network


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

Hotspot


Dive into the research topics where Dennys E. Cintra is active.

Publication


Featured researches published by Dennys E. Cintra.


The Journal of Neuroscience | 2009

Saturated Fatty Acids Produce an Inflammatory Response Predominantly through the Activation of TLR4 Signaling in Hypothalamus: Implications for the Pathogenesis of Obesity

Marciane Milanski; Giovanna R. Degasperi; Andressa Coope; Joseane Morari; Raphael G.P. Denis; Dennys E. Cintra; Daniela Miti Tsukumo; Gabriel F. Anhê; Maria do Carmo Estanislau do Amaral; Hilton Kenji Takahashi; Rui Curi; Helena C. F. Oliveira; José B.C. Carvalheira; Silvana Bordin; Mario J.A. Saad; Lício A. Velloso

In animal models of diet-induced obesity, the activation of an inflammatory response in the hypothalamus produces molecular and functional resistance to the anorexigenic hormones insulin and leptin. The primary events triggered by dietary fats that ultimately lead to hypothalamic cytokine expression and inflammatory signaling are unknown. Here, we test the hypothesis that dietary fats act through the activation of toll-like receptors 2/4 and endoplasmic reticulum stress to induce cytokine expression in the hypothalamus of rodents. According to our results, long-chain saturated fatty acids activate predominantly toll-like receptor 4 signaling, which determines not only the induction of local cytokine expression but also promotes endoplasmic reticulum stress. Rats fed on a monounsaturated fat-rich diet do not develop hypothalamic leptin resistance, whereas toll-like receptor 4 loss-of-function mutation and immunopharmacological inhibition of toll-like receptor 4 protects mice from diet-induced obesity. Thus, toll-like receptor 4 acts as a predominant molecular target for saturated fatty acids in the hypothalamus, triggering the intracellular signaling network that induces an inflammatory response, and determines the resistance to anorexigenic signals.


PLOS ONE | 2009

High-fat diet induces apoptosis of hypothalamic neurons.

Juliana C. Moraes; Andressa Coope; Joseane Morari; Dennys E. Cintra; Erika A. Roman; José Rodrigo Pauli; Talita Romanatto; José B.C. Carvalheira; Alexandre Leite Rodrigues de Oliveira; Mario J.A. Saad; Lício A. Velloso

Consumption of dietary fats is amongst the most important environmental factors leading to obesity. In rodents, the consumption of fat-rich diets blunts leptin and insulin anorexigenic signaling in the hypothalamus by a mechanism dependent on the in situ activation of inflammation. Since inflammatory signal transduction can lead to the activation of apoptotic signaling pathways, we evaluated the effect of high-fat feeding on the induction of apoptosis of hypothalamic cells. Here, we show that consumption of dietary fats induce apoptosis of neurons and a reduction of synaptic inputs in the arcuate nucleus and lateral hypothalamus. This effect is dependent upon diet composition, and not on caloric intake, since pair-feeding is not sufficient to reduce the expression of apoptotic markers. The presence of an intact TLR4 receptor, protects cells from further apoptotic signals. In diet-induced inflammation of the hypothalamus, TLR4 exerts a dual function, on one side activating pro-inflammatory pathways that play a central role in the development of resistance to leptin and insulin, and on the other side restraining further damage by controlling the apoptotic activity.


PLOS ONE | 2012

Unsaturated fatty acids revert diet-induced hypothalamic inflammation in obesity.

Dennys E. Cintra; Eduardo R. Ropelle; Juliana C. Moraes; José Rodrigo Pauli; Joseane Morari; Cláudio T. De Souza; Renato Grimaldi; Marcela Stahl; José B.C. Carvalheira; Mario J.A. Saad; Lício A. Velloso

Background In experimental models, hypothalamic inflammation is an early and determining factor in the installation and progression of obesity. Pharmacological and gene-based approaches have proven efficient in restraining inflammation and correcting the obese phenotypes. However, the role of nutrients in the modulation of hypothalamic inflammation is unknown. Methodology/Principal Findings Here we show that, in a mouse model of diet-induced obesity, partial substitution of the fatty acid component of the diet by flax seed oil (rich in C18:3) or olive oil (rich in C18:1) corrects hypothalamic inflammation, hypothalamic and whole body insulin resistance, and body adiposity. In addition, upon icv injection in obese rats, both ω3 and ω9 pure fatty acids reduce spontaneous food intake and body mass gain. These effects are accompanied by the reversal of functional and molecular hypothalamic resistance to leptin/insulin and increased POMC and CART expressions. In addition, both, ω3 and ω9 fatty acids inhibit the AMPK/ACC pathway and increase CPT1 and SCD1 expression in the hypothalamus. Finally, acute hypothalamic injection of ω3 and ω9 fatty acids activate signal transduction through the recently identified GPR120 unsaturated fatty acid receptor. Conclusions/Significance Unsaturated fatty acids can act either as nutrients or directly in the hypothalamus, reverting diet-induced inflammation and reducing body adiposity. These data show that, in addition to pharmacological and genetic approaches, nutrients can also be attractive candidates for controlling hypothalamic inflammation in obesity.


PLOS Biology | 2010

IL-6 and IL-10 Anti-Inflammatory Activity Links Exercise to Hypothalamic Insulin and Leptin Sensitivity through IKKβ and ER Stress Inhibition

Eduardo R. Ropelle; Marcelo B.S. Flores; Dennys E. Cintra; Guilherme Z. Rocha; José Rodrigo Pauli; Joseane Morari; Cláudio T. De Souza; Juliana C. Moraes; Patrícia O. Prada; Dioze Guadagnini; Rodrigo Miguel Marin; Alexandre G. Oliveira; Taize M. Augusto; Hernandes F. Carvalho; Lício A. Velloso; Mario J.A. Saad; José B.C. Carvalheira

Physical activity confers beneficial metabolic effects by inducing anti-inflammatory activity in the hypothalamus region of the brain in rodents, resulting in a reorganization of the set point of nutritional balance and reduced insulin and leptin resistance.


The Journal of Physiology | 2006

Reversal of diet-induced insulin resistance with a single bout of exercise in the rat: the role of PTP1B and IRS-1 serine phosphorylation

Eduardo R. Ropelle; José Rodrigo Pauli; Patrícia O. Prada; Cláudio T. De Souza; Paty K. Picardi; Marcel C. Faria; Dennys E. Cintra; Maria Fernanda A. Fernandes; Marcelo B.S. Flores; Lício A. Velloso; Mario J.A. Saad; José B.C. Carvalheira

Lifestyle interventions including exercise programmes are cornerstones in the prevention of obesity‐related diabetes. In this study, we demonstrate that a single bout of exercise inhibits high‐fat diet‐induced insulin resistance. Diet‐induced obesity (DIO) increased the expression and activity of the protein tyrosine phosphatase 1B (PTP1B) and attenuated insulin signalling in gastrocnemius muscle of rats, a phenomenon which was reversed by a single session of exercise. In addition, DIO was observed to lead to serine phosphorylation of insulin receptor substrate 1 (IRS‐1), which was also reversed by exercise in muscle in parallel with a reduction in c‐Jun N‐terminal kinase (JNK) activity. Thus, acute exercise increased the insulin sensitivity during high‐fat feeding in obese rats. Overall, these results provide new insights into the mechanism by which exercise restores insulin sensitivity.


Journal of Hepatology | 2008

Interleukin-10 is a protective factor against diet-induced insulin resistance in liver☆

Dennys E. Cintra; José Rodrigo Pauli; Eliana P. Araújo; Juliana C. Moraes; Cláudio T. De Souza; Marciane Milanski; Joseane Morari; Alessandra Gambero; Mario J.A. Saad; Lício A. Velloso

BACKGROUND/AIMS The anti-inflammatory cytokine, interleukin-10 (IL-10), is known to exert a protective role in hepatic damage caused by viruses, alcohol, autoimmunity and a number of experimental aggressors. Recently, a protective role for IL-10 has also been proposed in diet-induced hepatic dysfunction. However, studies about the mechanisms involved in this process are controversial. The objective of this study was to evaluate the role of endogenous IL-10 in the development of hepatic insulin resistance, associated with diet-induced fatty liver disease. METHODS Male Swiss mice treated for eight weeks with a high-fat diet became diabetic and developed non-alcoholic fatty liver disease, which is characterized by increased hepatic fat deposition and liver infiltration by F4/80 positive cells. This was accompanied by an increased hepatic expression of TNF-alpha, IL-6, IL-1beta and IL-10, and by an impaired insulin signal transduction through the insulin receptor/IRS1-IRS2/PI3-kinase/Akt/FOXO1 signaling pathway. RESULTS Upon endogenous IL-10 inhibition for 5 days, using two distinct methods, a neutralizing anti-IL-10 antibody and an antisense oligonucleotide against IL-10, increased hepatic expression of the inflammatory markers TNF-alpha, IL-6, IL-1beta and F4/80 was observed. This was accompanied by a significant negative modulation of insulin signal transduction through insulin receptor/IRS1-IRS2/PI3-kinase/Akt/FOXO1, and by the stimulation of hepatic signaling proteins involved in gluconeogenesis and lipid synthesis. CONCLUSIONS Thus, in an animal model of diet-induced fatty liver disease, the inhibition of IL-10 promotes the increased expression of inflammatory cytokines, the worsening of insulin signaling and the activation of gluconeogenic and lipidogenic pathways.


The Journal of Physiology | 2008

Acute physical exercise reverses S-nitrosation of the insulin receptor, insulin receptor substrate 1 and protein kinase B/Akt in diet-induced obese Wistar rats

José Rodrigo Pauli; Eduardo R. Ropelle; Dennys E. Cintra; Marco Antonio Carvalho-Filho; Juliana C. Moraes; Cláudio T. De Souza; Lício A. Velloso; José B.C. Carvalheira; Mario J.A. Saad

Early evidence demonstrates that exogenous nitric oxide (NO) and the NO produced by inducible nitric oxide synthase (iNOS) can induce insulin resistance. Here, we investigated whether this insulin resistance, mediated by S‐nitrosation of proteins involved in early steps of the insulin signal transduction pathway, could be reversed by acute physical exercise. Rats on a high‐fat diet were subjected to swimming for two 3 h‐long bouts, separated by a 45 min rest period. Two or 16 h after the exercise protocol the rats were killed and proteins from the insulin signalling pathway were analysed by immunoprecipitation and immunoblotting. We demonstrated that a high‐fat diet led to an increase in the iNOS protein level and S‐nitrosation of insulin receptor β (IRβ), insulin receptor substrate 1 (IRS1) and Akt. Interestingly, an acute bout of exercise reduced iNOS expression and S‐nitrosation of proteins involved in the early steps of insulin action, and improved insulin sensitivity in diet‐induced obesity rats. Furthermore, administration of GSNO (NO donor) prevents this improvement in insulin action and the use of an inhibitor of iNOS (l‐N6‐(1‐iminoethyl)lysine; l‐NIL) simulates the effects of exercise on insulin action, insulin signalling and S‐nitrosation of IRβ, IRS1 and Akt. In summary, a single bout of exercise reverses insulin sensitivity in diet‐induced obese rats by improving the insulin signalling pathway, in parallel with a decrease in iNOS expression and in the S‐nitrosation of IR/IRS1/Akt. The decrease in iNOS protein expression in the muscle of diet‐induced obese rats after an acute bout of exercise was accompanied by an increase in AMP‐activated protein kinase (AMPK) activity. These results provide new insights into the mechanism by which exercise restores insulin sensitivity.


Diabetes | 2011

Partial Reversibility of Hypothalamic Dysfunction and Changes in Brain Activity After Body Mass Reduction in Obese Subjects

Simone van de Sande-Lee; Fabricio Pereira; Dennys E. Cintra; Paula Teixeira Fernandes; Adilson R. Cardoso; Celia Regina Garlipp; Eliton A. Chaim; José Carlos Pareja; Bruno Geloneze; Li Min Li; Fernando Cendes; Lício A. Velloso

OBJECTIVE Inflammation and dysfunction of the hypothalamus are common features of experimental obesity. However, it is unknown whether obesity and massive loss of body mass can modify the immunologic status or the functional activity of the human brain. Therefore, the aim of this study was to determine the effect of body mass reduction on brain functionality. RESEARCH DESIGN AND METHODS In humans, changes in hypothalamic activity after a meal or glucose intake can be detected by functional magnetic resonance imaging (fMRI). Distinct fMRI analytic methods have been developed to explore changes in the brain’s activity in several physiologic and pathologic conditions. We used two analytic methods of fMRI to explore the changes in the brain activity after body mass reduction. RESULTS Obese patients present distinct functional activity patterns in selected brain regions compared with lean subjects. On massive loss of body mass, after bariatric surgery, increases in the cerebrospinal fluid (CSF) concentrations of interleukin (IL)-10 and IL-6 are accompanied by changes in fMRI patterns, particularly in the hypothalamus. CONCLUSIONS Massive reduction of body mass promotes a partial reversal of hypothalamic dysfunction and increases anti-inflammatory activity in the CSF.


Diabetes | 2009

EGFR Tyrosine Kinase Inhibitor (PD153035) Improves Glucose Tolerance and Insulin Action in High-Fat Diet–Fed Mice

Patrícia O. Prada; Eduardo R. Ropelle; Rosa H. Mourão; Cláudio T. De Souza; José Rodrigo Pauli; Dennys E. Cintra; André Almeida Schenka; Silvana A. Rocco; Roberto Rittner; Kleber G. Franchini; José Vassallo; Lício A. Velloso; José B.C. Carvalheira; Mario J.A. Saad

OBJECTIVE In obesity, an increased macrophage infiltration in adipose tissue occurs, contributing to low-grade inflammation and insulin resistance. Epidermal growth factor receptor (EGFR) mediates both chemotaxis and proliferation in monocytes and macrophages. However, the role of EGFR inhibitors in this subclinical inflammation has not yet been investigated. We investigated, herein, in vivo efficacy and associated molecular mechanisms by which PD153035, an EGFR tyrosine kinase inhibitor, improved diabetes control and insulin action. RESEARCH DESIGN AND METHODS The effect of PD153035 was investigated on insulin sensitivity, insulin signaling, and c-Jun NH2-terminal kinase (JNK) and nuclear factor (NF)-κB activity in tissues of high-fat diet (HFD)-fed mice and also on infiltration and the activation state of adipose tissue macrophages (ATMs) in these mice. RESULTS PD153035 treatment for 1 day decreased the protein expression of inducible nitric oxide synthase, tumor necrosis factor (TNF)-α, and interleukin (IL)-6 in the stroma vascular fraction, suggesting that this drug reduces the M1 proinflammatory state in ATMs, as an initial effect, in turn reducing the circulating levels of TNF-α and IL-6, and initiating an improvement in insulin signaling and sensitivity. After 14 days of drug administration, there was a marked improvement in glucose tolerance; a reduction in insulin resistance; a reduction in macrophage infiltration in adipose tissue and in TNF-α, IL-6, and free fatty acids; accompanied by an improvement in insulin signaling in liver, muscle, and adipose tissue; and also a decrease in insulin receptor substrate-1 Ser307 phosphorylation in JNK and inhibitor of NF-κB kinase (IKKβ) activation in these tissues. CONCLUSIONS Treatment with PD153035 improves glucose tolerance, insulin sensitivity, and signaling and reduces subclinical inflammation in HFD-fed mice.


PLOS ONE | 2008

Central Exercise Action Increases the AMPK and mTOR Response to Leptin

Eduardo R. Ropelle; Maria Fernanda A. Fernandes; Marcelo B.S. Flores; Mirian Ueno; Silvana A. Rocco; Rodrigo Miguel Marin; Dennys E. Cintra; Lício A. Velloso; Kleber G. Franchini; Mario J.A. Saad; José B.C. Carvalheira

AMP-activated protein kinase (AMPK) and mammalian Target of Rapamycin (mTOR) are key regulators of cellular energy balance and of the effects of leptin on food intake. Acute exercise is associated with increased sensitivity to the effects of leptin on food intake in an IL-6-dependent manner. To determine whether exercise ameliorates the AMPK and mTOR response to leptin in the hypothalamus in an IL-6-dependent manner, rats performed two 3-h exercise bouts, separated by one 45-min rest period. Intracerebroventricular IL-6 infusion reduced food intake and pretreatment with AMPK activators and mTOR inhibitor prevented IL-6-induced anorexia. Activators of AMPK and fasting increased food intake in control rats to a greater extent than that observed in exercised ones, whereas inhibitor of AMPK had the opposite effect. Furthermore, the reduction of AMPK and ACC phosphorylation and increase in phosphorylation of proteins involved in mTOR signal transduction, observed in the hypothalamus after leptin infusion, were more pronounced in both lean and diet-induced obesity rats after acute exercise. Treatment with leptin reduced food intake in exercised rats that were pretreated with vehicle, although no increase in responsiveness to leptin-induced anorexia after pretreatment with anti-IL6 antibody, AICAR or Rapamycin was detected. Thus, the effects of leptin on the AMPK/mTOR pathway, potentiated by acute exercise, may contribute to appetite suppressive actions in the hypothalamus.

Collaboration


Dive into the Dennys E. Cintra's collaboration.

Top Co-Authors

Avatar

José Rodrigo Pauli

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Eduardo R. Ropelle

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cláudio T. De Souza

Universidade do Extremo Sul Catarinense

View shared research outputs
Top Co-Authors

Avatar

Leandro Pereira de Moura

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Lício A. Velloso

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mario J.A. Saad

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Rafael Calais Gaspar

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Vitor Rosetto Muñoz

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge