Network


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

Hotspot


Dive into the research topics where Carlos K. Katashima is active.

Publication


Featured researches published by Carlos K. Katashima.


Embo Molecular Medicine | 2015

Alzheimer‐associated Aβ oligomers impact the central nervous system to induce peripheral metabolic deregulation

Julia R. Clarke; Natalia M. Lyra e Silva; Cláudia P. Figueiredo; Rudimar Luiz Frozza; José Henrique Ledo; Danielle Beckman; Carlos K. Katashima; Daniela S. Razolli; Bruno M. Carvalho; Renata Frazão; Marina Silveira; Felipe C. Ribeiro; Theresa R. Bomfim; Fernanda S. Neves; William L. Klein; Rodrigo Medeiros; Frank M. LaFerla; José B.C. Carvalheira; Mario J.A. Saad; Douglas P. Munoz; Lício A. Velloso; Sergio T. Ferreira; Fernanda G. De Felice

Alzheimers disease (AD) is associated with peripheral metabolic disorders. Clinical/epidemiological data indicate increased risk of diabetes in AD patients. Here, we show that intracerebroventricular infusion of AD‐associated Aβ oligomers (AβOs) in mice triggered peripheral glucose intolerance, a phenomenon further verified in two transgenic mouse models of AD. Systemically injected AβOs failed to induce glucose intolerance, suggesting AβOs target brain regions involved in peripheral metabolic control. Accordingly, we show that AβOs affected hypothalamic neurons in culture, inducing eukaryotic translation initiation factor 2α phosphorylation (eIF2α‐P). AβOs further induced eIF2α‐P and activated pro‐inflammatory IKKβ/NF‐κB signaling in the hypothalamus of mice and macaques. AβOs failed to trigger peripheral glucose intolerance in tumor necrosis factor‐α (TNF‐α) receptor 1 knockout mice. Pharmacological inhibition of brain inflammation and endoplasmic reticulum stress prevented glucose intolerance in mice, indicating that AβOs act via a central route to affect peripheral glucose homeostasis. While the hypothalamus has been largely ignored in the AD field, our findings indicate that AβOs affect this brain region and reveal novel shared molecular mechanisms between hypothalamic dysfunction in metabolic disorders and AD.


Diabetes | 2013

Targeted Disruption of Inducible Nitric Oxide Synthase Protects Against Aging, S -Nitrosation, and Insulin Resistance in Muscle of Male Mice

Eduardo R. Ropelle; José Rodrigo Pauli; Dennys E. Cintra; Adelino Silva; Cláudio T. De Souza; Dioze Guadagnini; Bruno M. Carvalho; Andrea M. Caricilli; Carlos K. Katashima; Marco Antonio Carvalho-Filho; Sandro M. Hirabara; Rui Curi; Lício A. Velloso; Mario J.A. Saad; José B.C. Carvalheira

Accumulating evidence has demonstrated that S-nitrosation of proteins plays a critical role in several human diseases. Here, we explored the role of inducible nitric oxide synthase (iNOS) in the S-nitrosation of proteins involved in the early steps of the insulin-signaling pathway and insulin resistance in the skeletal muscle of aged mice. Aging increased iNOS expression and S-nitrosation of major proteins involved in insulin signaling, thereby reducing insulin sensitivity in skeletal muscle. Conversely, aged iNOS-null mice were protected from S-nitrosation–induced insulin resistance. Moreover, pharmacological treatment with an iNOS inhibitor and acute exercise reduced iNOS-induced S-nitrosation and increased insulin sensitivity in the muscle of aged animals. These findings indicate that the insulin resistance observed in aged mice is mainly mediated through the S-nitrosation of the insulin-signaling pathway.


FEBS Open Bio | 2013

Diet-induced obesity induces endoplasmic reticulum stress and insulin resistance in the amygdala of rats

Gisele Castro; Maria Fernanda Condes Areias; Laís Weissmann; Paula G.F. Quaresma; Carlos K. Katashima; Mario J.A. Saad; Patrícia O. Prada

Insulin acts in the hypothalamus, decreasing food intake (FI) by the IR/PI3K/Akt pathway. This pathway is impaired in obese animals and endoplasmic reticulum (ER) stress and low‐grade inflammation are possible mechanisms involved in this impairment. Here, we highlighted the amygdala as an important brain region for FI regulation in response to insulin. This regulation was dependent on PI3K/AKT pathway similar to the hypothalamus. Insulin was able to decrease neuropeptide Y (NPY) and increase oxytocin mRNA levels in the amygdala via PI3K, which may contribute to hypophagia. Additionally, obese rats did not reduce FI in response to insulin and AKT phosphorylation was decreased in the amygdala, suggesting insulin resistance. Insulin resistance was associated with ER stress and low‐grade inflammation in this brain region. The inhibition of ER stress with PBA reverses insulin action/signaling, decreases NPY and increases oxytocin mRNA levels in the amygdala from obese rats, suggesting that ER stress is probably one of the mechanisms that induce insulin resistance in the amygdala.


American Journal of Physiology-endocrinology and Metabolism | 2013

Acute exercise suppresses hypothalamic PTP1B protein level and improves insulin and leptin signaling in obese rats

Eloize C. Chiarreotto-Ropelle; Luciana Santos Souza Pauli; Carlos K. Katashima; Gustavo Duarte Pimentel; Paty K. Picardi; Vagner Ramon Rodrigues Silva; Cláudio T. De Souza; Patrícia O. Prada; Dennys E. Cintra; José B.C. Carvalheira; Eduardo R. Ropelle; José Rodrigo Pauli

Hypothalamic inflammation is associated with insulin and leptin resistance, hyperphagia, and obesity. In this scenario, hypothalamic protein tyrosine phosphatase 1B (PTP1B) has emerged as the key phosphatase induced by inflammation that is responsible for the central insulin and leptin resistance. Here, we demonstrated that acute exercise reduced inflammation and PTP1B protein level/activity in the hypothalamus of obese rodents. Exercise disrupted the interaction between PTP1B with proteins involved in the early steps of insulin (IRβ and IRS-1) and leptin (JAK2) signaling, increased the tyrosine phosphorylation of these molecules, and restored the anorexigenic effects of insulin and leptin in obese rats. Interestingly, the anti-inflammatory action and the reduction of PTP1B activity mediated by exercise occurred in an interleukin-6 (IL-6)-dependent manner because exercise failed to reduce inflammation and PTP1B protein level after the disruption of hypothalamic-specific IL-6 action in obese rats. Conversely, intracerebroventricular administration of recombinant IL-6 reproduced the effects of exercise, improving hypothalamic insulin and leptin action by reducing the inflammatory signaling and PTP1B activity in obese rats at rest. Taken together, our study reports that physical exercise restores insulin and leptin signaling, at least in part, by reducing hypothalamic PTP1B protein level through the central anti-inflammatory response.


Nature Communications | 2014

Hypothalamic S1P/S1PR1 axis controls energy homeostasis

Vagner Silva; Thayana O. Micheletti; Gustavo Duarte Pimentel; Carlos K. Katashima; Luciene Lenhare; Joseane Morari; Maria Carolina Santos Mendes; Daniela S. Razolli; Guilherme Z. Rocha; Cláudio T. De Souza; Dongryeol Ryu; Patrícia O. Prada; Lício A. Velloso; José B.C. Carvalheira; José Rodrigo Pauli; Dennys E. Cintra; Eduardo R. Ropelle

Sphingosine 1-phosphate receptor 1 (S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/S1PR1/STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/S1PR1/STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats.


The Journal of Physiology | 2014

Exercise training decreases mitogen-activated protein kinase phosphatase-3 expression and suppresses hepatic gluconeogenesis in obese mice

Luciana Santos Souza Pauli; Eloize Cristina Chiarreotto Ropelle; Cláudio T. De Souza; Dennys E. Cintra; Adelino Sanchez Ramos da Silva; Bárbara de Almeida Rodrigues; Leandro Pereira de Moura; Rodolfo Marinho; Vanessa de Oliveira; Carlos K. Katashima; José Rodrigo Pauli; Eduardo R. Ropelle

When the hepatic insulin signaling is compromised, there is an inadequate suppression of gluconeogenic pathways, leading the organism to high levels of glucose. Studies with animals with obesity induced by high fat diet or genetically modified showed increased MKP‐3 expression and MKP‐3/Foxo1 association in liver, with a consequent increase in blood glucose concentration, development of insulin resistance and DM2. As a non‐pharmacological strategy recognized and indicated for prevention and treatment of diabetes is the regular practice of physical exercise. In this study we demostrated that physical training is an important tool capable of reducing insulin resistance in the liver by reducing the inflammatory process, including the inhibition of MKP‐3 and, therefore, suppress gluconeogenic program in obesity rats. The understanding of these new mechanisms by which physical training regulates glucose homeostasis has critical importance to health professionals for the understanding and prevention of diabetes.


Experimental Gerontology | 2017

Physical exercise increases Sestrin 2 protein levels and induces autophagy in the skeletal muscle of old mice

Luciene Lenhare; Barbara M. Crisol; Vagner Silva; Carlos K. Katashima; André V. Cordeiro; Karina D. Pereira; Augusto D. Luchessi; Adelino Sanchez Ramos da Silva; Dennys E. Cintra; Leandro Pereira de Moura; José Rodrigo Pauli; Eduardo R. Ropelle

ABSTRACT Sestrins and autophagy deficiencies are associated with several aging‐related organic dysfunctions and metabolic disorders. Here we evaluate the effects of acute exercise on Sestrin 2 (Sesn2) protein content and autophagy markers in the skeletal muscle of experimental models of aging. Twenty‐four months‐old C57BL/6 J male mice were submitted to a single bout of swimming exercise and the gastrocnemius muscle was evaluated by Western blot. Transcriptomic and phenotypic analysis were also performed by using strains of genetically‐diverse BXD mice. The bioinformatics analysis showed a negative correlation between Sesn2 mRNA levels in the skeletal muscle and body weight gain, plasma triglycerides and fasting glucose and positive correlation with several autophagic markers in the muscle of BXD mice. Consistent with these findings, low levels of Sesn2 protein content were observed in the gastrocnemius muscle of C57BL/6 J old mice when compared to young group. Interestingly, the acute aerobic exercise induced Sesn2 accumulation and modulated several markers of autophagy in the gastrocnemius muscle old mice, including unc‐51‐like kinase‐1 (Ulk1) phosphorylation and the protein levels of Atg5, Atg7, p62 and LC3‐II. Finally, exercise increased insulin sensitivity in old animals, as demonstrated by kITT. Taken together, these findings demonstrated the acutely, aerobic physical exercise recovers Sestrin 2 protein content and induces autophagy in the skeletal muscle of old mice, contributing with the improvement of insulin sensitivity an aging animal model. HIGHLIGHTSDownregulation of Sestrin2 in the skeletal muscle of old mice.Exercise stimulates Sestrin2 accumulation in the skeletal muscle of old mice.Physical exercise elicits autophagy in the skeletal muscle of old mice.


Obesity Reviews | 2017

Ursolic acid and mechanisms of actions on adipose and muscle tissue: a systematic review

Carlos K. Katashima; Vagner Silva; Tatyanne L. Gomes; Claude Pichard; Gustavo Duarte Pimentel

This systematic review aimed at addressing the ursolic acid actions as an adjunctive treatment of the obesity‐mediated metabolic abnormalities. To explore our aims, we used the literature search including clinical and animal studies using the Medline and Google Scholar (up to December 2015). Out of 63 screened studies, 17 presented eligibility criteria, such as the use of ursolic acid on adiposity, energy expenditure and skeletal muscle mass in mice and humans. In the literature, we found that several physiological and molecular mechanisms are implicated in the effects of ursolic acid on obesity, energy expenditure, hepatic steatosis, skeletal muscle mass loss and physical fitness, such as (1) increase of thermogenesis by modulation adipocyte transcription factors, activation of 5′ adenosine monophosphate‐activated protein kinase and overexpression of the uncoupling protein 1 thermogenic marker; (2) enhancement of skeletal muscle mass by activation in bloodstream growth hormone and insulin‐like growth factor‐1 concentrations secretion, as well as in the activation of mammalian target of rapamycin and inhibition of ring‐finger protein‐1; and (3) improvement of physical fitness by skeletal muscle proliferator‐activated receptor gamma co‐activator alpha and sirtuin 1 expression. Therefore, supplementation with ursolic acid may be an adjunctive therapy for prevention and treatment of obesity‐mediated and muscle mass‐mediated metabolic consequences.


Scientific Reports | 2017

iNOS promotes hypothalamic insulin resistance associated with deregulation of energy balance and obesity in rodents

Carlos K. Katashima; Vagner Silva; Luciene Lenhare; Rodrigo Miguel Marin; José B.C. Carvalheira

Inducible nitric oxide (iNOS)-mediated S-nitrosation of the metabolic signaling pathway has emerged as a post-translational modification that triggers insulin resistance in obesity and aging. However, the effects of S-nitrosation in controlling energy homeostasis are unknown. Thus, in the present study we aimed to evaluate the effects of S-nitrosation in insulin signaling pathway in the hypothalamus of rodents. Herein, we demonstrated that the intracerebroventricular infusion of the nitric oxide (NO) donor S-nitrosoglutathione (GSNO) promoted hypothalamic insulin signaling resistance and replicated the food intake pattern of obese individuals. Indeed, obesity induced S-nitrosation of hypothalamic IR and Akt, whereas inhibition of iNOS or S-nitrosation of insulin signaling pathway protected against hypothalamic insulin resistance and normalized energy homeostasis. Overall, these findings indicated that S-nitrosation of insulin signaling pathway is required to sustain hypothalamic insulin resistance in obesity.


Aging-us | 2016

Hypothalamic S1P/S1PR1 axis controls energy homeostasis in Middle-Aged Rodents: the reversal effects of physical exercise

Vagner Silva; Carlos K. Katashima; Carla Grazielle Bueno Silva; Luciene Lenhare; Thayana O. Micheletti; Rafael Ludemann Camargo; Ana C. Ghezzi; Juliana A. Camargo; Alexandre Moura Assis; Natália Tobar; Joseane Morari; Daniela S. Razolli; Leandro Pereira de Moura; José Rodrigo Pauli; Dennys E. Cintra; Lício A. Velloso; Mario J.A. Saad; Eduardo R. Ropelle

Recently, we demonstrated that the hypothalamic S1PR1/STAT3 axis plays a critical role in the control of food consumption and energy expenditure in rodents. Here, we found that reduction of hypothalamic S1PR1 expression occurs in an age-dependent manner, and was associated with defective thermogenic signaling and weight gain. To address the physiological relevance of these findings, we investigated the effects of chronic and acute exercise on the hypothalamic S1PR1/STAT3 axis. Chronic exercise increased S1PR1 expression and STAT3 phosphorylation in the hypothalamus, restoring the anorexigenic and thermogenic signals in middle-aged mice. Acutely, exercise increased sphingosine-1-phosphate (S1P) levels in the cerebrospinal fluid (CSF) of young rats, whereas the administration of CSF from exercised young rats into the hypothalamus of middle-aged rats at rest was sufficient to reduce the food intake. Finally, the intracerebroventricular (ICV) administration of S1PR1 activators, including the bioactive lipid molecule S1P, and pharmacological S1PR1 activator, SEW2871, induced a potent STAT3 phosphorylation and anorexigenic response in middle-aged rats. Overall, these results suggest that hypothalamic S1PR1 is important for the maintenance of energy balance and provide new insights into the mechanism by which exercise controls the anorexigenic and thermogenic signals in the central nervous system during the aging process.

Collaboration


Dive into the Carlos K. Katashima's collaboration.

Top Co-Authors

Avatar

Eduardo R. Ropelle

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Vagner Silva

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

Dennys E. Cintra

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Luciene Lenhare

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lício A. Velloso

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Leandro Pereira de Moura

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
Researchain Logo
Decentralizing Knowledge