Network


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

Hotspot


Dive into the research topics where Andressa Coope is active.

Publication


Featured researches published by Andressa Coope.


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.


web science | 2012

Inhibition of Hypothalamic Inflammation Reverses Diet-Induced Insulin Resistance in the Liver

Marciane Milanski; Ana Paula Arruda; Andressa Coope; Letícia M. Ignacio-Souza; Carla E. Nunez; Erika A. Roman; Talita Romanatto; Lívia Bitencourt Pascoal; Andrea M. Caricilli; Marcio Alberto Torsoni; Patrícia O. Prada; Mario J.A. Saad; Lício A. Velloso

Defective liver gluconeogenesis is the main mechanism leading to fasting hyperglycemia in type 2 diabetes, and, in concert with steatosis, it is the hallmark of hepatic insulin resistance. Experimental obesity results, at least in part, from hypothalamic inflammation, which leads to leptin resistance and defective regulation of energy homeostasis. Pharmacological or genetic disruption of hypothalamic inflammation restores leptin sensitivity and reduces adiposity. Here, we evaluate the effect of a hypothalamic anti-inflammatory approach to regulating hepatic responsiveness to insulin. Obese rodents were treated by intracerebroventricular injections, with immunoneutralizing antibodies against Toll-like receptor (TLR)4 or tumor necrosis factor (TNF)α, and insulin signal transduction, hepatic steatosis, and gluconeogenesis were evaluated. The inhibition of either TLR4 or TNFα reduced hypothalamic inflammation, which was accompanied by the reduction of hypothalamic resistance to leptin and improved insulin signal transduction in the liver. This was accompanied by reduced liver steatosis and reduced hepatic expression of markers of steatosis. Furthermore, the inhibition of hypothalamic inflammation restored defective liver glucose production. All these beneficial effects were abrogated by vagotomy. Thus, the inhibition of hypothalamic inflammation in obesity results in improved hepatic insulin signal transduction, leading to reduced steatosis and reduced gluconeogenesis. All these effects are mediated by parasympathetic signals delivered by the vagus nerve.


FEBS Letters | 2008

AdipoR1 mediates the anorexigenic and insulin/leptin-like actions of adiponectin in the hypothalamus

Andressa Coope; Marciane Milanski; Eliana P. Araújo; Marcos Antonio Tambascia; Mario J.A. Saad; Bruno Geloneze; Lício A. Velloso

Adiponectin exerts an insulin‐sensitizing effect, improving insulin action in peripheral tissues and restraining insulin resistance. Here, we explore the hypothesis that adiponectin can reproduce some of the actions of insulin/leptin in the hypothalamus. The presence of AdipoR1 and AdipoR2 was mapped to the arcuate and lateral hypothalamic nuclei. Icv adiponectin reduced food intake, which was accompanied by activation/engagement of IRS1/2, ERK, Akt, FOXO1, JAK2 and STAT3. All these actions were dependent on AdipoR1, since inhibition of this receptor, and not of AdipoR2, completely reversed the effects described above. Thus, adiponectin acts in the hypothalamus, activating elements of the canonical insulin and leptin signaling pathways and promoting reduction of food intake.


Endocrinology | 2011

Low-Grade Hypothalamic Inflammation Leads to Defective Thermogenesis, Insulin Resistance, and Impaired Insulin Secretion

Ana Paula Arruda; Marciane Milanski; Andressa Coope; Adriana Souza Torsoni; Eduardo R. Ropelle; Denise P. Carvalho; José B.C. Carvalheira; Lício A. Velloso

Hypothalamic inflammation is present in animal models of obesity, and the intracerebroventricular injection of TNFα can reproduce a number of features of the hypothalamus of obese animals. Because obesity is a risk factor for type 2 diabetes (DM2) we hypothesized that, by inducing hypothalamic inflammation, we could reproduce some clinical features of DM2. Lean Wistar rats and TNF receptor 1-knockout mice were employed to determine the effects of hypothalamic actions of TNFα on thermogenesis and metabolic parameters. Signal transduction and protein expression were evaluated by immunoblot and real-time PCR. Thermogenesis was evaluated in living rats, and respirometry was determined in isolated muscle fiber. In Wistar rats, hypothalamic TNFα blunts the anorexigenic effect of leptin, which is accompanied by reduced leptin signaling and increased expression of suppressor of cytokine signaling 3. In addition, hypothalamic TNFα reduces O(2) consumption and the expression of thermogenic proteins in brown adipose tissue and skeletal muscle. Furthermore, hypothalamic inflammation increases base-line plasma insulin and insulin secretion by isolated pancreatic islets, which is accompanied by an impaired insulin signal transduction in liver and skeletal muscle. Hypothalamic inflammation induced by stearic acid also reduces O(2) consumption and blunts peripheral insulin signal transduction. The use of intracerebroventricular infliximab restores O(2) consumption in obese rats, whereas TNF receptor 1-knockout mice are protected from diet-induced reduced thermogenesis and defective insulin signal transduction. Thus, low-grade inflammation of the hypothalamus is sufficient to induce changes in a number of parameters commonly impaired in obesity and DM2, and TNFα is an important mediator of this process.


Endocrinology | 2010

Hypothalamic Actions of Tumor Necrosis Factor α Provide the Thermogenic Core for the Wastage Syndrome in Cachexia

Ana Paula Arruda; Marciane Milanski; Talita Romanatto; Carina Solon; Andressa Coope; Luciane C. Alberici; William T. Festuccia; Sandro M. Hirabara; Eduardo R. Ropelle; Rui Curi; José B.C. Carvalheira; Anibal E. Vercesi; Lício A. Velloso

TNFalpha is an important mediator of catabolism in cachexia. Most of its effects have been characterized in peripheral tissues, such as skeletal muscle and fat. However, by acting directly in the hypothalamus, TNFalpha can activate thermogenesis and modulate food intake. Here we show that high concentration TNFalpha in the hypothalamus leads to increased O(2) consumption/CO(2) production, increased body temperature, and reduced caloric intake, resulting in loss of body mass. Most of the thermogenic response is produced by beta 3-adrenergic signaling to the brown adipose tissue (BAT), leading to increased BAT relative mass, reduction in BAT lipid quantity, and increased BAT mitochondria density. The expression of proteins involved in BAT thermogenesis, such as beta 3-adrenergic receptor, peroxisomal proliferator-activated receptor-gamma coactivator-1 alpha, and uncoupling protein-1, are increased. In the hypothalamus, TNFalpha produces reductions in neuropeptide Y, agouti gene-related peptide, proopiomelanocortin, and melanin-concentrating hormone, and increases CRH and TRH. The activity of the AMP-activated protein kinase signaling pathway is also decreased in the hypothalamus of TNFalpha-treated rats. Upon intracerebroventricular infliximab treatment, tumor-bearing and septic rats present a significantly increased survival. In addition, the systemic inhibition of beta 3-adrenergic signaling results in a reduced body mass loss and increased survival in septic rats. These data suggest hypothalamic TNFalpha action to be important mediator of the wastage syndrome in cachexia.


Neuroscience | 2010

TNF-α transiently induces endoplasmic reticulum stress and an incomplete unfolded protein response in the hypothalamus

Raphael G.P. Denis; Ana Paula Arruda; Talita Romanatto; Marciane Milanski; Andressa Coope; Carina Solon; Daniela S. Razolli; Lício A. Velloso

In diet-induced obesity, hypothalamic inflammation is triggered as an outcome of prolonged exposure to dietary fats. Toll-like receptor 4 (TLR4) activation plays a central role in this process, inducing endoplasmic reticulum stress and activating inflammatory cytokine gene transcription. Although saturated fatty acids can induce endoplasmic reticulum stress in the hypothalamus, it is unknown whether inflammatory cytokines alone can activate this mechanism. Here, rats were treated with TNF-α or lyposaccharide (LPS) and endoplasmic reticulum stress and unfolded protein response were evaluated by immunoblot and polymerase chain reaction (PCR). Activation of TLR4 by LPS was capable of inducing a complete endoplasmic reticulum stress and unfolded protein response through the PERK/eIF2α and IRE1α/XBP1 pathways. Conversely, TNF-α, injected either locally or systemically, was unable to induce a complete program of unfolded protein response, although the activation of endoplasmic reticulum stress was achieved to a certain degree. Thus, in the hypothalamus, the isolated action of TNF-α is insufficient to produce the activation of a complete program of unfolded protein response.


Journal of Biological Chemistry | 2012

Chaperone insufficiency links TLR4 protein signaling to endoplasmic reticulum stress.

Andressa Coope; Marciane Milanski; Ana Paula Arruda; Letícia M. Ignacio-Souza; Mario J.A. Saad; Gabriel F. Anhê; Lício A. Velloso

Background: Activation of TLR4 leads to endoplasmic reticulum stress. However, the mechanisms involved in this phenomenon are unknown. Results: In TLR4 signaling, insufficient GRP94 and GRP78 mediate the activation of endoplasmic reticulum stress. Conclusion: The insufficiency of chaperone expression links TLR4 signaling to endoplasmic reticulum stress. Significance: This study may improve our understanding about the inflammatory response in metabolic and infectious disease. Inflammation plays an important pathogenic role in a number of metabolic diseases such as obesity, type 2 diabetes, and atherosclerosis. The activation of inflammation in these diseases depends at least in part on the combined actions of TLR4 signaling and endoplasmic reticulum stress, which by acting in concert can boost the inflammatory response. Defining the mechanisms involved in this phenomenon may unveil potential targets for the treatment of metabolic/inflammatory diseases. Here we used LPS to induce endoplasmic reticulum stress in the human monocyte cell-line, THP-1. The unfolded protein response, produced after LPS, was dependent on CD14 activity but not on RNA-dependent protein kinase and could be inhibited by an exogenous chemical chaperone. The induction of the endoplasmic reticulum resident chaperones, GRP94 and GRP78, by LPS was of a much lower magnitude than the effect of LPS on TLR4 and MD-2 expression. In face of this apparent insufficiency of chaperone expression, we induced the expression of GRP94 and GRP78 by glucose deprivation. This approach completely reverted endoplasmic reticulum stress. The inhibition of either GRP94 or GRP78 with siRNA was sufficient to rescue the protective effect of glucose deprivation on LPS-induced endoplasmic reticulum stress. Thus, insufficient LPS-induced chaperone expression links TLR4 signaling to endoplasmic reticulum stress.


European Journal of Endocrinology | 2016

MECHANISMS IN ENDOCRINOLOGY: Metabolic and inflammatory pathways on the pathogenesis of type 2 diabetes

Andressa Coope; Adriana Souza Torsoni; Lício A. Velloso

Obesity is the main risk factor for type 2 diabetes (T2D). Studies performed over the last 20 years have identified inflammation as the most important link between these two diseases. During the development of obesity, there is activation of subclinical inflammatory activity in tissues involved in metabolism and energy homeostasis. Intracellular serine/threonine kinases activated in response to inflammatory factors can catalyse the inhibitory phosphorylation of key proteins of the insulin-signalling pathway, leading to insulin resistance. Moreover, during the progression of obesity and insulin resistance, the pancreatic islets are also affected by inflammation, contributing to β-cell failure and leading to the onset of T2D. In this review, we will present the main mechanisms involved in the activation of obesity-associated metabolic inflammation and discuss potential therapeutic opportunities that can be developed to treat obesity-associated metabolic diseases.


PLOS ONE | 2015

Saturated Fatty Acids Modulate Autophagy's Proteins in the Hypothalamus

Mariana Portovedo; Letícia M. Ignacio-Souza; Bruna Bombassaro; Andressa Coope; Andressa Reginato; Daniela S. Razolli; Marcio Alberto Torsoni; Adriana Souza Torsoni; Raquel Franco Leal; Lício A. Velloso; Marciane Milanski

Autophagy is an important process that regulates cellular homeostasis by degrading dysfunctional proteins, organelles and lipids. In this study, the hypothesis that obesity could lead to impairment in hypothalamic autophagy in mice was evaluated by examining the hypothalamic distribution and content of autophagic proteins in animal with obesity induced by 8 or 16 weeks high fat diet to induce obesity and in response to intracerebroventricular injections of palmitic acid. The results showed that chronic exposure to a high fat diet leads to an increased expression of inflammatory markers and downregulation of autophagic proteins. In obese mice, autophagic induction leads to the downregulation of proteins, such as JNK and Bax, which are involved in the stress pathways. In neuron cell- line, palmitate has a direct effect on autophagy even without inflammatory activity. Understanding the cellular and molecular bases of overnutrition is essential for identifying new diagnostic and therapeutic targets for obesity.

Collaboration


Dive into the Andressa Coope's collaboration.

Top Co-Authors

Avatar

Lício A. Velloso

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Marciane Milanski

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Raquel Franco Leal

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

João José Fagundes

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mariana Portovedo

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Ana Paula Arruda

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
Researchain Logo
Decentralizing Knowledge