Network


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

Hotspot


Dive into the research topics where Mario J.A. Saad is active.

Publication


Featured researches published by Mario J.A. Saad.


Diabetes | 2007

Loss-of-Function Mutation in Toll-Like Receptor 4 Prevents Diet-Induced Obesity and Insulin Resistance

Daniela Miti Tsukumo; Marco Antonio Carvalho-Filho; José B.C. Carvalheira; Patrícia O. Prada; Sandro M. Hirabara; André Almeida Schenka; Eliana P. Araújo; José Vassallo; Rui Curi; Lício A. Velloso; Mario J.A. Saad

Obesity is associated with insulin resistance and a state of abnormal inflammatory response. The Toll-like receptor (TLR)4 has an important role in inflammation and immunity, and its expression has been reported in most tissues of the body, including the insulin-sensitive ones. Because it is activated by lipopolysaccharide and saturated fatty acids, which are inducers of insulin resistance, TLR4 may be a candidate for participation in the cross-talk between inflammatory and metabolic signals. Here, we show that C3H/HeJ mice, which have a loss-of-function mutation in TLR4, are protected against the development of diet-induced obesity. In addition, these mice demonstrate decreased adiposity, increased oxygen consumption, a decreased respiratory exchange ratio, improved insulin sensitivity, and enhanced insulin-signaling capacity in adipose tissue, muscle, and liver compared with control mice during high-fat feeding. Moreover, in these tissues, control mice fed a high-fat diet show an increase in IκB kinase complex and c-Jun NH2-terminal kinase activity, which is prevented in C3H/HeJ mice. In isolated muscles from C3H/HeJ mice, protection from saturated fatty acid–induced insulin resistance is observed. Thus, TLR4 appears to be an important mediator of obesity and insulin resistance and a potential target for the therapy of these highly prevalent medical conditions.


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 Biology | 2011

Gut Microbiota Is a Key Modulator of Insulin Resistance in TLR 2 Knockout Mice

Andrea M. Caricilli; Paty K. Picardi; Lélia L. de Abreu; Mirian Ueno; Patrícia O. Prada; Eduardo R. Ropelle; Sandro M. Hirabara; Ângela Castoldi; Pedro Vieira; Niels Olsen Saraiva Camara; Rui Curi; José B.C. Carvalheira; Mario J.A. Saad

A genetic and pharmacological approach reveals novel insights into how changes in gut microbiota can subvert genetically predetermined phenotypes from lean to obese.


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.


Diabetes | 2008

A Central Role for Neuronal AMP-Activated Protein Kinase (AMPK) and Mammalian Target of Rapamycin (mTOR) in High-Protein Diet–Induced Weight Loss

Eduardo R. Ropelle; José Rodrigo Pauli; Maria Fernanda A. Fernandes; Silvana A. Rocco; Rodrigo Miguel Marin; Joseane Morari; Kellen K. Souza; Marília M. Dias; Maria Cristina Cintra Gomes-Marcondes; José Antonio Rocha Gontijo; Kleber G. Franchini; Lício A. Velloso; Mario J.A. Saad; José B.C. Carvalheira

OBJECTIVE—A high-protein diet (HPD) is known to promote the reduction of body fat, but the mechanisms underlying this change are unclear. AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) function as majors regulators of cellular metabolism that respond to changes in energy status, and recent data demonstrated that they also play a critical role in systemic energy balance. Here, we sought to determine whether the response of the AMPK and mTOR pathways could contribute to the molecular effects of an HPD. RESEARCH DESIGN AND METHODS—Western blotting, confocal microscopy, chromatography, light microscopy, and RT-PCR assays were combined to explore the anorexigenic effects of an HPD. RESULTS—An HPD reduced food intake and induced weight loss in both normal rats and ob/ob mice. The intracerebroventricular administration of leucine reduced food intake, and the magnitude of weight loss and reduction of food intake in a leucine-supplemented diet are similar to that achieved by HPD in normal rats and in ob/ob mice, suggesting that leucine is a major component of the effects of an HPD. Leucine and HPD decrease AMPK and increase mTOR activity in the hypothalamus, leading to inhibition of neuropeptide Y and stimulation of pro-opiomelanocortin expression. Consistent with a cross-regulation between AMPK and mTOR to control food intake, our data show that the activation of these enzymes occurs in the same specific neuronal subtypes. CONCLUSIONS—These findings provide support for the hypothesis that AMPK and mTOR interact in the hypothalamus to regulate feeding during HPD in a leucine-dependent manner.


Clinical Cancer Research | 2011

Metformin Amplifies Chemotherapy-Induced AMPK Activation and Antitumoral Growth

Guilherme Z. Rocha; Marília M. Dias; Eduardo R. Ropelle; Felipe Osório-Costa; Franco A. Rossato; Anibal E. Vercesi; Mario J.A. Saad; José B.C. Carvalheira

Purpose: Metformin is a widely used antidiabetic drug whose anticancer effects, mediated by the activation of AMP-activated protein kinase (AMPK) and reduction of mTOR signaling, have become noteworthy. Chemotherapy produces genotoxic stress and induces p53 activity, which can cross-talk with AMPK/mTOR pathway. Herein, we investigate whether the combination of metformin and paclitaxel has an effect in cancer cell lines. Experimental Design: Human tumors were xenografted into severe combined immunodeficient (SCID) mice and the cancer cell lines were treated with only paclitaxel or only metformin, or a combination of both drugs. Western blotting, flow cytometry, and immunohistochemistry were then used to characterize the effects of the different treatments. Results: The results presented herein show that the addition of metformin to paclitaxel leads to quantitative potentialization of molecular signaling through AMPK and a subsequent potent inhibition of the mTOR signaling pathway. Treatment with metformin and paclitaxel resulted in an increase in the number of cells arrested in the G2–M phase of the cell cycle, and decreased the tumor growth and increased apoptosis in tumor-bearing mice, when compared with individual drug treatments. Conclusion: We have provided evidence for a convergence of metformin and paclitaxel induced signaling at the level of AMPK. This mechanism shows how different drugs may cooperate to augment antigrowth signals, and suggests that target activation of AMPK by metformin may be a compelling ally in cancer treatment. Clin Cancer Res; 17(12); 3993–4005. ©2011 AACR.


FEBS Letters | 2001

Insulin modulates leptin-induced STAT3 activation in rat hypothalamus

José B.C. Carvalheira; Rodrigo Marins Peixoto Siloto; Inara Ignacchitti; Sigisfredo L. Brenelli; Carla R.O. Carvalho; Adilson Leite; Lício A. Velloso; José Antonio Rocha Gontijo; Mario J.A. Saad

Insulin and leptin have overlapping effects in the control of energy homeostasis, but the molecular basis of this synergism is unknown. Insulin signals through a receptor tyrosine kinase that phosphorylates and activates the docking proteins IRSs (insulin receptor substrates), whereas the leptin receptor and its associated protein tyrosine kinase JAK2 (Janus kinase 2) mediate phosphorylation and activation of the transcription factor STAT3 (signal transducer and activator of transcription). Here, we present evidence for the integration of leptin and insulin signals in the hypothalamus. Insulin induced JAK2 tyrosine phosphorylation, leptin receptor phosphorylation which, in the presence of leptin, augmented the interaction between STAT3 and this receptor. Insulin also increased the leptin‐induced phosphorylation of STAT3 and its activation. These results indicate that insulin modulates the leptin signal transduction pathway, and may provide a molecular basis for the coordinated effects of insulin and leptin in feeding behavior and weight control.


American Journal of Physiology-endocrinology and Metabolism | 1998

Pinealectomy causes glucose intolerance and decreases adipose cell responsiveness to insulin in rats

Fabio Bessa Lima; Ubiratan Fabres Machado; Ione Bartol; Patricia Monteiro Seraphim; Doris Hissako Sumida; Solange Marta Franzói de Moraes; N.S. Hell; Maristela Mitiko Okamoto; Mario J.A. Saad; Carla R.O. Carvalho; José Cipolla-Neto

Although the pineal gland influences several physiological systems, only a few studies have investigated its role in the intermediary metabolism. In the present study, male Wistar rats, pinealectomized or sham-operated 6 wk before the experiment, were submitted to both intravenous glucose tolerance tests (IVGTT) and insulin binding as well as glucose transport assays in isolated adipocytes. The insulin receptor tyrosine kinase activity was assessed in liver and muscle. The insulin secretory response during the IVGTT was impaired, particularly in the afternoon, and the glucose transport responsiveness was 33% lower in pinealectomized rats. However, no difference was observed in the insulin receptor number of adipocytes between groups as well as in insulin-stimulated tyrosine kinase activity, indicating that the initial steps in the insulin signaling were well conserved. Conversely, a 40% reduction in adipose tissue GLUT-4 content was detected. In conclusion, pinealectomy is responsible for both impaired insulin secretion and action, emphasizing the influence of the pineal gland on glucose metabolism.

Collaboration


Dive into the Mario J.A. Saad's collaboration.

Top Co-Authors

Avatar

Lício A. Velloso

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrícia O. Prada

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Dioze Guadagnini

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Eduardo R. Ropelle

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Carla R.O. Carvalho

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

José Rodrigo Pauli

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Cláudio T. De Souza

Universidade do Extremo Sul Catarinense

View shared research outputs
Top Co-Authors

Avatar

Eliana P. Araújo

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Mirian Ueno

State University of Campinas

View shared research outputs
Researchain Logo
Decentralizing Knowledge