Network


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

Hotspot


Dive into the research topics where Antonio C. Boschero is active.

Publication


Featured researches published by Antonio C. Boschero.


Journal of Biological Chemistry | 2009

Deletion of tumor necrosis factor-alpha-receptor 1 (TNFR1) protects against diet-induced obesity by means of increased thermogenesis

Talita Romanatto; Erika A. Roman; Ana Paula Arruda; Raphael G.P. Denis; Carina Solon; Marciane Milanski; Juliana C. Moraes; Maria Lúcia Bonfleur; Giovanna R. Degasperi; Paty K. Picardi; Sandro M. Hirabara; Antonio C. Boschero; Rui Curi; Lício A. Velloso

In diet-induced obesity, hypothalamic and systemic inflammatory factors trigger intracellular mechanisms that lead to resistance to the main adipostatic hormones, leptin and insulin. Tumor necrosis factor-α (TNF-α) is one of the main inflammatory factors produced during this process and its mechanistic role as an inducer of leptin and insulin resistance has been widely investigated. Most of TNF-α inflammatory signals are delivered by TNF receptor 1 (R1); however, the role played by this receptor in the context of obesity-associated inflammation is not completely known. Here, we show that TNFR1 knock-out (TNFR1 KO) mice are protected from diet-induced obesity due to increased thermogenesis. Under standard rodent chow or a high-fat diet, TNFR1 KO gain significantly less body mass despite increased caloric intake. Visceral adiposity and mean adipocyte diameter are reduced and blood concentrations of insulin and leptin are lower. Protection from hypothalamic leptin resistance is evidenced by increased leptin-induced suppression of food intake and preserved activation of leptin signal transduction through JAK2, STAT3, and FOXO1. Under the high-fat diet, TNFR1 KO mice present a significantly increased expression of the thermogenesis-related neurotransmitter, TRH. Further evidence of increased thermogenesis includes increased O2 consumption in respirometry measurements, increased expressions of UCP1 and UCP3 in brown adipose tissue and skeletal muscle, respectively, and increased O2 consumption by isolated skeletal muscle fiber mitochondria. This demonstrates that TNF-α signaling through TNFR1 is an important mechanism involved in obesity-associated defective thermogenesis.


Pflügers Archiv: European Journal of Physiology | 1978

The stimulus secretion coupling of glucose-induced insulin release. XXVII. Effect of glucose on K+ fluxes in isolated islets.

Willy Malaisse; Antonio C. Boschero; Shoji Kawazu; John-Charles Hutton

The effect of glucose upon the handling of K+ by islets of Langerhans removed from normal rats was investigated by measuring both the net uptake of86Rb+ and its efflux from prelabelled islets. The inflow of K+ into islet cells is mediated, in part at least, by an ouabain-sensitive pump. Glucose fails to affect the inflow rate of K+, but it apparently decreases the permeability of islet cells plasma membrane to effluent K+. The glucose-induced change in permeability is a rapid and rapidly reversible phenomenon. Under steady-state conditions, it leads to an increase in the islet cells K+ pool and a decrease of its fractional turnover rate.


Hormone Research in Paediatrics | 1977

Calcium antagonists and islet function. XI. Effect of nifedipine.

Willy Malaisse; Antonio C. Boschero

Nifedipine inhibits calcium-45 net uptake and glucose-induced insulin release in rat pancreatic islets, without affecting glucose oxidation and calcium-45 efflux. These findings are compatible with the view that nifedipine inhibits the entry of calcium in the B cell, by interfering with the plasma membrane ionophoretic channels.


PLOS ONE | 2012

Altered Glucose Homeostasis and Hepatic Function in Obese Mice Deficient for Both Kinin Receptor Genes

Carlos C. Barros; Anderson Sola Haro; F.J. Russo; Ines Schadock; Sandro Soares de Almeida; Rosane A. Ribeiro; Emerielle C. Vanzela; Valéria Pereira Lanzoni; Flavio C. Barros; Milton Rocha Moraes; Marcelo A. Mori; Reury Frank Pereira Bacurau; Martin Würtele; Antonio C. Boschero; Everardo M. Carneiro; Michael Bader; João Bosco Pesquero; Ronaldo C. Araujo

The Kallikrein-Kinin System (KKS) has been implicated in several aspects of metabolism, including the regulation of glucose homeostasis and adiposity. Kinins and des-Arg-kinins are the major effectors of this system and promote their effects by binding to two different receptors, the kinin B2 and B1 receptors, respectively. To understand the influence of the KKS on the pathophysiology of obesity and type 2 diabetes (T2DM), we generated an animal model deficient for both kinin receptor genes and leptin (obB1B2KO). Six-month-old obB1B2KO mice showed increased blood glucose levels. Isolated islets of the transgenic animals were more responsive to glucose stimulation releasing greater amounts of insulin, mainly in 3-month-old mice, which was corroborated by elevated serum C-peptide concentrations. Furthermore, they presented hepatomegaly, pronounced steatosis, and increased levels of circulating transaminases. This mouse also demonstrated exacerbated gluconeogenesis during the pyruvate challenge test. The hepatic abnormalities were accompanied by changes in the gene expression of factors linked to glucose and lipid metabolisms in the liver. Thus, we conclude that kinin receptors are important for modulation of insulin secretion and for the preservation of normal glucose levels and hepatic functions in obese mice, suggesting a protective role of the KKS regarding complications associated with obesity and T2DM.


The FASEB Journal | 2007

Inhibition of UCP2 expression reverses diet-induced diabetes mellitus by effects on both insulin secretion and action

Cláudio T. De Souza; Eliana P. Araújo; Luiz F. Stoppiglia; José Rodrigo Pauli; Eduardo R. Ropelle; Silvana A. Rocco; Rodrigo Miguel Marin; Kleber G. Franchini; José B.C. Carvalheira; Mario J.A. Saad; Antonio C. Boschero; Everardo M. Carneiro; Lício A. Velloso

Recent characterization of the ability of uncoupling protein 2 (UCP2) to reduce ATP production and inhibit insulin secretion by pancreatic β‐cells has placed this mitochondrial protein as a candidate target for therapeutics in diabetes mellitus. In the present study we evaluate the effects of short‐term treatment of two animal models of type 2 diabetes mellitus with an antisense oligonucleotide to UCP2. In both models, Swiss mice (made obese and diabetic by a hyperlipidic diet) and ob/ob mice, the treatment resulted in a significant improvement in the hyperglyce‐mic syndrome. This effect was due not only to an improvement of insulin secretion, but also to improved peripheral insulin action. In isolated pancreatic islets, the partial inhibition of UCP2 increased ATP content, followed by increased glucose‐stimulated insulin secretion. This was not accompanied by increased expression of enzymes involved in protection against oxida‐tive stress. The evaluation of insulin action in peripheral tissues revealed that the inhibition of UCP2 expression significantly improved insulin signal trans‐duction in adipose tissue. In conclusion, short‐term inhibition of UCP2 expression ameliorates the hyper‐glycemic syndrome in two distinct animal models of obesity and diabetes. Metabolic improvement is due to a combined effect on insulin‐producing pancreatic islets and in at least one peripheral tissue that acts as a target for insulin.—De Souza, C. T., Araújo, E. P., Stoppiglia, L. F., Pauli, J. R., Ropelle, E., Rocco, S. A., Marin, R. M., Franchini, K. G., Carvalheira, J. B., Saad, M. J., Boschero, A. C., Carneiro, E. M., Velloso, L. A. Inhibition of UCP2 expression reverses diet‐induced diabetes mellitus by effects on both insulin secretion and action. FASEB J. 21, 1153–1163 (2007)


Journal of Biological Chemistry | 2011

Inflammation of the hypothalamus leads to defective pancreatic islet function.

Vivian C. Calegari; Adriana Souza Torsoni; Ennerielle C. Vanzela; Eliana P. Araújo; Joseane Morari; Claudio C. Zoppi; Lourenço Sbragia; Antonio C. Boschero; Lício A. Velloso

Type 2 diabetes mellitus results from the complex association of insulin resistance and pancreatic β-cell failure. Obesity is the main risk factor for type 2 diabetes mellitus, and recent studies have shown that, in diet-induced obesity, the hypothalamus becomes inflamed and dysfunctional, resulting in the loss of the perfect coupling between caloric intake and energy expenditure. Because pancreatic β-cell function is, in part, under the control of the autonomic nervous system, we evaluated the role of hypothalamic inflammation in pancreatic islet function. In diet-induced obesity, the earliest markers of hypothalamic inflammation are present at 8 weeks after the beginning of the high fat diet; similarly, the loss of the first phase of insulin secretion is detected at the same time point and is restored following sympathectomy. Intracerebroventricular injection of a low dose of tumor necrosis factor α leads to a dysfunctional increase in insulin secretion and activates the expression of a number of markers of apoptosis in pancreatic islets. In addition, the injection of stearic acid intracerebroventricularly, which leads to hypothalamic inflammation through the activation of tau-like receptor-4 and endoplasmic reticulum stress, produces an impairment of insulin secretion, accompanied by increased expression of markers of apoptosis. The defective insulin secretion, in this case, is partially dependent on sympathetic signal-induced peroxisome proliferator receptor-γ coactivator Δα and uncoupling protein-2 expression and is restored after sympathectomy or following PGC1α expression inhibition by an antisense oligonucleotide. Thus, the autonomic signals generated in concert with hypothalamic inflammation can impair pancreatic islet function, a phenomenon that may explain the early link between obesity and defective insulin secretion.


FEBS Letters | 1995

Glucose- and insulin-induced phosphorylation of the insulin receptor and its primary substrates IRS-1 and IRS-2 in rat pancreatic islets.

Lício A. Velloso; E.M. Carneiro; S.C. Crepaldi; Antonio C. Boschero; M. J. A. Saad

The presence of tyrosine‐phospborylated proteins was studied in cultured rat pancreatic islets. Immunoblotting performed with total extracts of islets cultured in the presence of 1.8 or 5.6 mM glucose revealed at least three distinct tyrosinephos‐phosphorylated bands (25 kDa, 95 kDa and 165–185 kDa). After 12 h incubation in medium containing 1.8 mM glucose, a pulse exposition to 11 or 22 mM glucose or to 10−7 M insulin led to a substantial increase in the phosphorylation of all three bands, with no appearance of novel bands. Immunoprecipitation with specific antibodies demonstrated that the signal detected at 95 kDa corresponds to the β subunit of the insulin receptor (IR) while the band at 165–185 kDa corresponds to the early substrates of the insulin receptor, IRS‐1 and IRS‐2. Immunoprecipitation with IRS‐1 or IRS‐2 antisera detected their association with the lipid metabolizing enzyme phosphatidylinositol 3‐kinase (PI 3‐kinase). Thus, this is the first demonstration that elements involved in the insulin‐signalling pathway of traditional target tissues are also present in pancreatic islets and are potentially involved in auto‐ and paracrine‐signalling in this organ.


American Journal of Physiology-endocrinology and Metabolism | 2009

High doses of dexamethasone induce increased beta-cell proliferation in pancreatic rat islets

Alex Rafacho; Tânia M. Cestari; Sebastião Roberto Taboga; Antonio C. Boschero; José Roberto Bosqueiro

Activation of insulin signaling and cell cycle intermediates is required for adult beta-cell proliferation. Here, we report a model to study beta-cell proliferation in living rats by administering three different doses of dexamethasone (0.1, 0.5, and 1.0 mg/kg ip, DEX 0.1, DEX 0.5, and DEX 1.0, respectively) for 5 days. Insulin sensitivity, insulin secretion, and histomorphometric data were investigated. Western blotting was used to analyze the levels of proteins related to the control of beta-cell growth. DEX 1.0 rats, which present moderate hyperglycemia and marked hyperinsulinemia, exhibited a 5.1-fold increase in beta-cell proliferation and an increase (17%) in beta-cell size, with significant increase in beta-cell mass, compared with control rats. The hyperinsulinemic but euglycemic DEX 0.5 rats also showed a significant 3.6-fold increase in beta-cell proliferation. However, DEX 0.1 rats, which exhibited the lowest degree of insulin resistance, compensate for insulin demand by improving only islet function. Activation of the insulin receptor substrate 2/phosphatidylinositol 3-kinase/serine-threonine kinase/ribosomal protein S6 kinase pathway, as well as protein retinoblastoma in islets from DEX 1.0 and DEX 0.5, but not in DEX 0.1, rats was also observed. Therefore, increasing doses of dexamethasone induce three different degrees of insulin requirement in living rats, serving as a model to investigate compensatory beta-cell alterations. Augmented beta-cell mass involves beta-cell hyperplasia and, to a lower extent, beta-cell hypertrophy. We suggest that alterations in circulating insulin and, to a lesser extent, glucose levels could be the major stimuli for beta-cell proliferation in the dexamethasone-induced insulin resistance.


Journal of Clinical Investigation | 1978

The stimulus-secretion coupling of glucose-induced insulin release. Metabolic and functional effects of NH4+ in rat islets.

Abdullah Sener; John-Charles Hutton; Shoji Kawazu; Antonio C. Boschero; Guido Somers; Ghislain Devis; André Herchuelz; Willy Malaisse

NH4+ caused a dose-related, rapid, and reversible inhibition of glucose-stimulated insulin release by isolated rat islets. It also inhibited glyceraldehyde-, Ba2+-, and sulfonylurea-stimulated insulun secretion. NH4+ failed to affect glucose utilization and oxidation, glucose-stimulated proinsulin biosynthesis, the concentration of ATP, AD, and AMP, and the intracellular pH. NH4+ also failed to affect the ability of theophylline and cytochalasin B to augment glucose-induced insulin release. However, in the presence and absence of glucose, accumulation of NH4+ in islet cells was associated with a fall in the concentration of NADH and HADPH and a concomitant alteration of 86Rb+ and 45Ca2+ (or 133Ba2+) handling. These findings suggest that reduced pyridine nucleotides, generated by the metabolism of endogenous of exogenous nutrients, may modulate ionophoretic processes in the islet cells and by doing so, affect the net uptake of Ca2+ and subsequent release of insulin.


Diabetologia | 2003

Peroxisome proliferator-activated receptor γ coactivator-1-dependent uncoupling protein-2 expression in pancreatic islets of rats: a novel pathway for neural control of insulin secretion

C. T. De Souza; Alessandra L. Gasparetti; Márcio Pereira-da-Silva; Eliana P. Araújo; José B.C. Carvalheira; M. J. A. Saad; Antonio C. Boschero; Everardo M. Carneiro; Lício A. Velloso

Aims/hypothesisSympathetic inputs inhibit insulin secretion through α2-adrenergic receptors coupled with Gi protein. High adrenergic tonus generated by exposure of homeothermic animals to cold reduces insulin secretion. In this study we evaluate the participation of UCP-2 in cold-induced regulation of insulin secretion.MethodsStatic insulin secretion studies, western blotting and immunohistochemistry were used in this investigation.ResultsExposure of rats to cold during 8 days promoted 60% (n=15, p<0.05) reduction of basal serum insulin levels concentration accompanied by reduction of the area under insulin curve during i.p. GTT (50%, n=15, p<0.05). Isolated islets from cold-exposed rats secreted 57% (n=6, p<0.05) less insulin following a glucose challenge. Previous sympathectomy, partially prevented the effect of cold exposure upon insulin secretion. Islets isolated from cold-exposed rats expressed 51% (n=6, p<0.5) more UCP-2 than islets from control rats, while the inhibition of UCP-2 expression by antisense oligonucleotide treatment partially restored insulin secretion of islets obtained from cold-exposed rats. Cold exposure also induced an increase of 69% (n=6, p<0.05) in PGC-1 protein content in pancreatic islets. Inhibition of islet PGC-1 expression by antisense oligonucleotide abrogated cold-induced UCP-2 expression and partially restored insulin secretion in islets exposed to cold.Conclusion/interpreatationOur data indicate that sympathetic tonus generated by exposure of rats to cold induces the expression of PGC-1, which participates in the control of UCP-2 expression in pancreatic islets. Increased UCP-2 expression under these conditions could reduce the beta-cell ATP/ADP ratio and negatively regulate insulin secretion.

Collaboration


Dive into the Antonio C. Boschero's collaboration.

Top Co-Authors

Avatar

Everardo M. Carneiro

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Lício A. Velloso

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Luiz F. Rezende

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Márcia Q. Latorraca

Universidade Federal de Mato Grosso

View shared research outputs
Top Co-Authors

Avatar

Willy Malaisse

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Rosane Aparecida Ribeiro

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Thiago M. Batista

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Emerielle C. Vanzela

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Claudio C. Zoppi

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Mario J.A. Saad

State University of Campinas

View shared research outputs
Researchain Logo
Decentralizing Knowledge