Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Cell Physiol ; 234(1): 880-890, 2018 01.
Article in English | MEDLINE | ID: mdl-30078194

ABSTRACT

This study investigated the effects of exercise training in regulating inflammatory processes, endoplasmic reticulum stress, and apoptosis in hypothalamic neurons of obese mice. Swiss mice were distributed into three groups: Lean mice (Lean), sedentary animals fed a standard diet; obese mice (Obese), sedentary animals fed a high-fat diet (HFD); trained obese mice (T. Obese), animals fed with HFD and concurrently subjected to an endurance training protocol for 8 weeks. In the endurance training protocol, mice ran on a treadmill at 60% of peak workload for 1 hr, 5 days/week for 8 weeks. Twenty-four hours after the last exercise session, the euthanasia was performed. Western blot, quantitative real-time polymerase chain reaction, and terminal deoxynucleotide transferase biotin-dUTP nick end-labeling (TUNEL) techniques were used for the analysis of interest. The results show exercise training increased phosphorylation of leptin signaling pathway proteins (pJAK2/pSTAT3) and reduced the content of tumor necrosis factor α, toll-like receptor 4, suppressor of cytokine signaling 3, protein-tyrosine phosphatase 1B as well as the phosphorylation of IkB kinase in the hypothalamus of T. Obese animals. A reduction of macrophage activation and phosphorylation of eukaryotic initiation factor 2α, and protein kinase RNA-like endoplasmic reticulum kinase (PERK) were also observed in exercised animals. Furthermore, exercise decreased the expression of the proapoptotic protein (PARP1) and increased anti-inflammatory (IL-10) and antiapoptotic (Bcl2) proteins. Using the TUNEL technique, we observed that the exercised animals had lower DNA fragmentation. Finally, physical exercise preserved pro-opiomelanocortin messenger RNA content. In conclusion, exercise training was able to reorganize the control of the energy balance through anti-inflammatory and antiapoptotic responses in hypothalamic tissue of obese mice.


Subject(s)
Endurance Training , Inflammation/physiopathology , Obesity/therapy , Physical Conditioning, Animal , Animals , Apoptosis/genetics , Diet, High-Fat , Energy Metabolism/genetics , Gene Expression Regulation , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation/therapy , Interleukin-10/genetics , Mice , Mice, Obese , Neurons/metabolism , Neurons/pathology , Obesity/physiopathology , Poly (ADP-Ribose) Polymerase-1/genetics , Proto-Oncogene Proteins c-bcl-2/genetics
3.
Eur J Endocrinol ; 175(3): R97-R105, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27006108

ABSTRACT

Selected subpopulations of hypothalamic neurons play important roles in the regulation of whole body energy homeostasis. Studies have shown that the saturated fats present in large amounts in western diets can activate an inflammatory response in the hypothalamus, affecting the capacity of such neurons to respond appropriately to satiety and adipostatic signals. In the first part of this review, we will explore the mechanisms behind saturated fatty acid-induced hypothalamic dysfunction. Next, we will present and discuss recent studies that have identified the mechanisms that mediate some of the anti-inflammatory actions of unsaturated fatty acids in the hypothalamus and the potential for exploring these mechanisms to prevent or treat obesity.


Subject(s)
Hypothalamus/physiopathology , Inflammation/physiopathology , Obesity/physiopathology , Humans , Insulin Resistance/physiology , Neurons/physiology , Nutritional Status
4.
Endocrinology ; 156(1): 103-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25375037

ABSTRACT

The anomalous activation of toll-like receptor 4 (TLR4) by dietary fats is one of the most important mechanisms linking obesity to insulin resistance. TLR4 is expressed in most tissues of the body, but its activity in the cells of the immune system is expected to underlie its most important roles of inducing inflammation and insulin resistance. Here we explore the hypothesis that TLR4 expression in bone marrow-derived cells mediates most of the actions of this receptor as an inducer of insulin resistance. Wild type and TLR4-mutant mice were used in bone marrow transplant experiments producing chimeras that harbored the functional receptor in all cells of the body except bone marrow-derived cells or only in bone marrow-derived cells. Transplanted mice were fed chow or a high-fat diet, and glucose homeostasis was evaluated by glucose and insulin tolerance tests. Insulin signal transduction and the expression of markers of inflammation were evaluated in the liver and white adipose tissue. In addition, we performed liver histology and evaluated the expression of gluconeogenic enzymes. The expression of TLR4 in bone marrow-derived cells only, but not in non-bone marrow-derived tissues only, was a determining factor in the induction of diet-induced insulin resistance, which was accompanied by an increased expression of inflammatory markers in both white adipose tissue and liver as well as increased liver steatosis and increased expression of gluconeogenic enzymes. TLR4 expressed in bone marrow-derived cells is an important mediator of obesity-associated insulin resistance in mice.


Subject(s)
Dietary Fats/adverse effects , Gene Expression Regulation/physiology , Insulin Resistance/physiology , Macrophages/drug effects , Macrophages/metabolism , Toll-Like Receptor 4/metabolism , Adipose Tissue/metabolism , Animals , Bone Marrow Transplantation , Dietary Fats/administration & dosage , Inflammation/genetics , Inflammation/metabolism , Insulin Resistance/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Proto-Oncogene Proteins c-akt , Toll-Like Receptor 4/genetics
5.
Diabetes ; 63(11): 3770-84, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24947351

ABSTRACT

Hypothalamic inflammation is a common feature of experimental obesity. Dietary fats are important triggers of this process, inducing the activation of toll-like receptor-4 (TLR4) signaling and endoplasmic reticulum stress. Microglia cells, which are the cellular components of the innate immune system in the brain, are expected to play a role in the early activation of diet-induced hypothalamic inflammation. Here, we use bone marrow transplants to generate mice chimeras that express a functional TLR4 in the entire body except in bone marrow-derived cells or only in bone marrow-derived cells. We show that a functional TLR4 in bone marrow-derived cells is required for the complete expression of the diet-induced obese phenotype and for the perpetuation of inflammation in the hypothalamus. In an obesity-prone mouse strain, the chemokine CX3CL1 (fractalkine) is rapidly induced in the neurons of the hypothalamus after the introduction of a high-fat diet. The inhibition of hypothalamic fractalkine reduces diet-induced hypothalamic inflammation and the recruitment of bone marrow-derived monocytic cells to the hypothalamus; in addition, this inhibition reduces obesity and protects against diet-induced glucose intolerance. Thus, fractalkine is an important player in the early induction of diet-induced hypothalamic inflammation, and its inhibition impairs the induction of the obese and glucose intolerance phenotypes.


Subject(s)
Chemokine CX3CL1/metabolism , Hypothalamus/metabolism , Inflammation/metabolism , Obesity/metabolism , Animals , Chemokine CX3CL1/genetics , Diet, High-Fat/adverse effects , Flow Cytometry , Hypothalamus/immunology , Immunoblotting , Inflammation/etiology , Inflammation/immunology , Male , Mice , Obesity/etiology , Obesity/immunology , Real-Time Polymerase Chain Reaction , Signal Transduction , Toll-Like Receptor 4/metabolism
6.
Food Chem ; 138(1): 574-80, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23265526

ABSTRACT

This study proposes a FRAP assay adapted to FIA system with a merging zones configuration. The FIA system conditions were optimised with the response surface methodology using the central composite rotatable design. The optimisation parameters studied were: the carrier flow rate, the lengths of the sample and reagent loops, and reactor length. The conditions selected in accordance with the results were: carrier flow rate of 1.00 ml/min, length of the loops 18.2 cm and length of the reaction coil 210.1 cm. The detection and quantification limits were, respectively, 28.6 and 86.8 µmol/l Fe(2+), and the precision was 1.27%. The proposed method had an analytical frequency of 30 samples/h and about 95% less volume of FRAP reagent was consumed. The FRAP assay adapted to the FIA system under the optimised conditions was utilised to determine the antioxidant activity of tea samples.


Subject(s)
Antioxidants/analysis , Chemistry Techniques, Analytical/methods , Ferric Compounds/chemistry , Flow Injection Analysis/methods , Tea/chemistry , Oxidation-Reduction
7.
PLoS One ; 7(1): e30571, 2012.
Article in English | MEDLINE | ID: mdl-22279596

ABSTRACT

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.


Subject(s)
Dietary Fats, Unsaturated/administration & dosage , Hypothalamus/drug effects , Inflammation/prevention & control , Obesity/prevention & control , Animals , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Eating/drug effects , Fatty Acids/blood , Fatty Acids/chemistry , Fatty Acids/metabolism , Fatty Acids, Omega-3/administration & dosage , Gene Expression/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Immunoblotting , Inflammation/blood , Inflammation/etiology , Insulin Resistance , Male , Mice , Nerve Tissue Proteins/genetics , Obesity/etiology , Obesity/physiopathology , Olive Oil , Plant Oils/administration & dosage , Pro-Opiomelanocortin/genetics , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Weight Gain/drug effects
8.
J Environ Manage ; 92(1): 178-84, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20869158

ABSTRACT

Activated carbons were prepared from flamboyant pods by NaOH activation at three different NaOH:char ratios: 1:1 (AC-1), 2:1 (AC-2), and 3:1 (AC-3). The properties of these carbons, including BET surface area, pore volume, pore size distribution, and pore diameter, were characterized from N(2) adsorption isotherms. The activated carbons obtained were essentially microporous and had BET surface area ranging from 303 to 2463 m(2) g(-1).(13)C (CP/MAS and MAS) solid-state NMR shows that the lignocellulosic structures were completely transformed into a polycyclic material after activation process, thermogravimetry shows a high thermal resistance, Boehm titration and Fourier-transform infrared spectroscopy allowed characterizing the presence of functional groups on the surface of activated carbons. Scanning electron microscopy images showed a high pore development. The experimental results indicated the potential use of flamboyant pods as a precursor material in the preparation of activated carbon.


Subject(s)
Carbon/chemistry , Fabaceae/chemistry , Lignin/chemistry , Porosity , Seeds , Temperature
9.
PLoS Biol ; 8(8)2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20808781

ABSTRACT

Overnutrition caused by overeating is associated with insulin and leptin resistance through IKKbeta activation and endoplasmic reticulum (ER) stress in the hypothalamus. Here we show that physical exercise suppresses hyperphagia and associated hypothalamic IKKbeta/NF-kappaB activation by a mechanism dependent upon the pro-inflammatory cytokine interleukin (IL)-6. The disruption of hypothalamic-specific IL-6 action blocked the beneficial effects of exercise on the re-balance of food intake and insulin and leptin resistance. This molecular mechanism, mediated by physical activity, involves the anti-inflammatory protein IL-10, a core inhibitor of IKKbeta/NF-kappaB signaling and ER stress. We report that exercise and recombinant IL-6 requires IL-10 expression to suppress hyperphagia-related obesity. Moreover, in contrast to control mice, exercise failed to reverse the pharmacological activation of IKKbeta and ER stress in C3H/HeJ mice deficient in hypothalamic IL-6 and IL-10 signaling. Hence, inflammatory signaling in the hypothalamus links beneficial physiological effects of exercise to the central action of insulin and leptin.


Subject(s)
Anti-Inflammatory Agents/metabolism , Endoplasmic Reticulum/pathology , I-kappa B Proteins/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Physical Conditioning, Animal/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Energy Metabolism , Hyperphagia , Hypothalamus/physiopathology , Insulin/physiology , Interleukin-10/pharmacology , Interleukin-6/pharmacology , Leptin/physiology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Obesity/metabolism , Rats , Rats, Wistar
10.
Mech Ageing Dev ; 131(5): 323-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20307567

ABSTRACT

The insulin resistance associated with aging is improved by exercise, but the molecular mechanisms of this improvement are not fully understood. We investigated whether the improvement in insulin action, associated with acute exercise in old rats is dependent on the modulation of pIRS-1Ser307, JNK, IkBalpha and PTP-1B. Aging rats were subjected to swimming for two 1.5-h long bouts, separated by a 45min rest period. Sixteen hours after the exercise, the rats were killed and proteins from the insulin signaling pathway were analyzed by immunoblotting. Our results show that the reduction in glucose disappearance rate (Kitt), observed in aged rats, was restored at 16h after exercise. Aging led to an increase in Ser307 phosphorylation of IRS-1, and this was reversed by exercise in the skeletal muscle, in parallel with a reduction in pJNK and IkBalpha degradation. Moreover, aging induced an increase in the expression of PTP-1B and attenuated insulin signaling in the muscle of rats, a phenomenon that was reversed by exercise. Interestingly, the decrease in PTP-1B expression in the muscle of exercised old rats was accompanied by an increase in SIRT1 expression. These results provide new insights into the mechanisms by which exercise restores insulin sensitivity during aging.


Subject(s)
Aging/physiology , Insulin Resistance , Insulin/metabolism , Muscle, Skeletal/physiology , Physical Conditioning, Animal , Aging/metabolism , Animals , I-kappa B Proteins/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins/metabolism , MAP Kinase Kinase 4/metabolism , Male , Muscle, Skeletal/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Rats , Rats, Wistar , Serine/metabolism , Signal Transduction , Sirtuin 1/biosynthesis
11.
J Transl Med ; 8: 11, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20113505

ABSTRACT

BACKGROUND: Ileal pouch-anal anastomosis (IPAA) is the surgical procedure of choice for patients with refractory ulcerative colitis (UC) and for familial adenomatous polyposis (FAP) with many rectal polyps. Pouchitis is one of the more frequent complications after IPAA in UC patients; however, it is rare in FAP. OBJECTIVE: Evaluate pro-apoptotic activity in endoscopically and histological normal mucosa of the ileal pouch in patients with UC and FAP. METHODS: Eighteen patients (nine with UC and nine with FAP) with J pouch after total rectocolectomy were studied. Biopsies were obtained from the mucosa of the pouch and from normal ileum. The specimens were snap-frozen and the expressions of Bax and Bcl-2 were determined by immunoblot of protein extracts and by immunohistochemistry analysis. FADD, Caspase-8, APAF-1 and Caspase-9 were evaluated by immunoprecipitation and immunoblot. RESULTS: Patients with UC had significantly higher protein levels of Bax and APAF-1, Caspase-9 than patients with FAP, but were similar to controls. The expressions of Bcl-2 and FADD, Caspase-8 were similar in the groups. Immunohistochemistry for Bax showed less intensity of immunoreactions in FAP than in UC and Controls. Bcl-2 immunostaining was similar among the groups. CONCLUSION: Patients with FAP present lower levels of pro-apoptotic proteins in all methods applied, even in the absence of clinical and endoscopic pouchitis and dysplasia in the histological analysis. These findings may explain a tendency of up-regulation of apoptosis in UC patients, resulting in higher rates of progression to pouchitis in these patients, which could correlate with mucosal atrophy that occurs in inflamed tissue. However, FAP patients had low pro-apoptotic activity in the mucosa, and it could explain the tendency to low cell turn over and presence of adenomas in this syndrome.


Subject(s)
Adenomatous Polyposis Coli/surgery , Apoptosis/physiology , Colitis, Ulcerative/surgery , Colonic Pouches/pathology , Intestinal Mucosa/pathology , Adenomatous Polyposis Coli/pathology , Animals , Apoptosis Regulatory Proteins/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Colitis, Ulcerative/pathology , Colonic Pouches/adverse effects , Humans , Ileum/anatomy & histology , Ileum/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
12.
J Biol Chem ; 284(52): 36213-36222, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19858212

ABSTRACT

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-alpha (TNF-alpha) 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-alpha 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 O(2) consumption in respirometry measurements, increased expressions of UCP1 and UCP3 in brown adipose tissue and skeletal muscle, respectively, and increased O(2) consumption by isolated skeletal muscle fiber mitochondria. This demonstrates that TNF-alpha signaling through TNFR1 is an important mechanism involved in obesity-associated defective thermogenesis.


Subject(s)
Obesity/metabolism , Oxygen Consumption , Receptors, Tumor Necrosis Factor, Type I/metabolism , Thermogenesis , Tumor Necrosis Factor-alpha/metabolism , Abdominal Fat/metabolism , Adipose Tissue, Brown/metabolism , Animals , Diet/adverse effects , Dietary Fats/adverse effects , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Inflammation/genetics , Inflammation/metabolism , Insulin/metabolism , Ion Channels/metabolism , Janus Kinase 2/metabolism , Leptin/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Obesity/genetics , Rats , Receptors, Tumor Necrosis Factor, Type I/genetics , STAT3 Transcription Factor/metabolism , Uncoupling Protein 1 , Uncoupling Protein 3
13.
PLoS One ; 4(4): e5045, 2009.
Article in English | MEDLINE | ID: mdl-19340313

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Dietary Fats/administration & dosage , Hypothalamus/drug effects , Neurons/drug effects , Animals , Dietary Fats/pharmacology , Hypothalamus/cytology , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C3H , Microscopy, Electron, Transmission , Neurons/cytology , Polymerase Chain Reaction , Rats
14.
J Physiol ; 587(Pt 10): 2341-51, 2009 05 15.
Article in English | MEDLINE | ID: mdl-19332486

ABSTRACT

Insulin signalling in the hypothalamus plays a role in maintaining body weight. The forkhead transcription factor Foxo1 is an important mediator of insulin signalling in the hypothalamus. Foxo1 stimulates the transcription of the orexigenic neuropeptide Y and Agouti-related protein through the phosphatidylinositol-3-kinase/Akt signalling pathway, but the role of hypothalamic Foxo1 in insulin resistance and obesity remains unclear. Here, we identify that a high-fat diet impaired insulin-induced hypothalamic Foxo1 phosphorylation and degradation, increasing the nuclear Foxo1 activity and hyperphagic response in rats. Thus, we investigated the effects of the intracerebroventricular (i.c.v.) microinfusion of Foxo1-antisense oligonucleotide (Foxo1-ASO) and evaluated the food consumption and weight gain in normal and diet-induced obese (DIO) rats. Three days of Foxo1-ASO microinfusion reduced the hypothalamic Foxo1 expression by about 85%. i.c.v. infusion of Foxo1-ASO reduced the cumulative food intake (21%), body weight change (28%), epididymal fat pad weight (22%) and fasting serum insulin levels (19%) and increased the insulin sensitivity (34%) in DIO but not in control animals. Collectively, these data showed that the Foxo1-ASO treatment blocked the orexigenic effects of Foxo1 and prevented the hyperphagic response in obese rats. Thus, pharmacological manipulation of Foxo1 may be used to prevent or treat obesity.


Subject(s)
Eating/drug effects , Forkhead Transcription Factors/metabolism , Gene Expression/drug effects , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Obesity/drug therapy , Oligonucleotides, Antisense/pharmacology , Adipose Tissue, White/anatomy & histology , Adipose Tissue, White/drug effects , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Diet , Energy Intake/drug effects , Epididymis/anatomy & histology , Epididymis/drug effects , Forkhead Transcription Factors/genetics , Hypothalamus/drug effects , Insulin/administration & dosage , Insulin/blood , Insulin/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Male , Nerve Tissue Proteins/genetics , Obesity/blood , Obesity/pathology , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptor, Insulin/metabolism , p300-CBP Transcription Factors/metabolism
15.
FEBS Lett ; 582(20): 3103-10, 2008 Sep 03.
Article in English | MEDLINE | ID: mdl-18703058

ABSTRACT

Uncoupling protein 2 (UCP2) is highly expressed in the hypothalamus; however, little is known about the functions it exerts in this part of the brain. Here, we hypothesized that UCP2 protects hypothalamic cells from oxidative and pro-apoptotic damage generated by inflammatory stimuli. Intracerebroventricular injection of tumor necrosis factor alpha (TNF-alpha)-induced an increase of UCP2 expression in the hypothalamus, which was accompanied by increased expression of markers of oxidative stress and pro-apoptotic proteins. The inhibition of UCP2 expression by an antisense oligonucleotide enhanced the damaging effects of TNF-alpha. Conversely, increasing the hypothalamic expression of UCP2 by cold exposure reversed most of the effects of the cytokine. Thus, UCP2 acts as a protective factor against cellular damage induced by an inflammatory stimulus in the hypothalamus.


Subject(s)
Apoptosis , Hypothalamus/cytology , Hypothalamus/metabolism , Ion Channels/physiology , Mitochondrial Proteins/physiology , Animals , Cells, Cultured , Cold Temperature , Ion Channels/antagonists & inhibitors , Ion Channels/biosynthesis , Male , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/biosynthesis , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology , Uncoupling Protein 2
16.
J Hepatol ; 48(4): 628-37, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18267346

ABSTRACT

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.


Subject(s)
Fatty Liver/metabolism , Insulin Resistance/physiology , Interleukin-10/metabolism , Liver/metabolism , Animals , Blood Glucose/metabolism , Disease Models, Animal , Disease Progression , Enzyme-Linked Immunosorbent Assay , Fatty Liver/pathology , Gene Expression , Glucose Tolerance Test , Immunoblotting , Immunoprecipitation , Insulin/blood , Interleukin-10/genetics , Interleukin-10/immunology , Liver/drug effects , Liver/pathology , Male , Mice , Oligonucleotides, Antisense/pharmacology , RNA/genetics , Signal Transduction/genetics , Thionucleotides/pharmacology
17.
J Physiol ; 586(2): 659-71, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17974582

ABSTRACT

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 beta (IR beta), 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 IR beta, 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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Obesity/metabolism , Physical Conditioning, Animal/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , AMP-Activated Protein Kinase Kinases , Adaptor Proteins, Signal Transducing/drug effects , Animals , Diet/adverse effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Insulin Receptor Substrate Proteins , Insulin Resistance/physiology , Lysine/analogs & derivatives , Lysine/pharmacology , Male , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/metabolism , Obesity/etiology , Protein Kinases/drug effects , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Rats , Rats, Wistar , Receptor, Insulin/drug effects , S-Nitrosoglutathione/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Peptides ; 28(5): 1050-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17459524

ABSTRACT

Acting in the hypothalamus, tumor necrosis factor-alpha (TNF-alpha) produces a potent anorexigenic effect. However, the molecular mechanisms involved in this phenomenon are poorly characterized. In this study, we investigate the capacity of TNF-alpha to activate signal transduction in the hypothalamus through elements of the pathways employed by the anorexigenic hormones insulin and leptin. High dose TNF-alpha promotes a reduction of 25% in 12h food intake, which is an inhibitory effect that is marginally inferior to that produced by insulin and leptin. In addition, high dose TNF-alpha increases body temperature and respiratory quotient, effects not reproduced by insulin or leptin. TNF-alpha, predominantly at the high dose, is also capable of activating canonical pro-inflammatory signal transduction in the hypothalamus, inducing JNK, p38, and NFkappaB, which results in the transcription of early responsive genes and expression of proteins of the SOCS family. Also, TNF-alpha activates signal transduction through JAK-2 and STAT-3, but does not activate signal transduction through early and intermediary elements of the insulin/leptin signaling pathways such as IRS-2, Akt, ERK and FOXO1. When co-injected with insulin or leptin, TNF-alpha, at both high and low doses, partially impairs signal transduction through IRS-2, Akt, ERK and FOXO1 but not through JAK-2 and STAT-3. This effect is accompanied by the partial inhibition of the anorexigenic effects of insulin and leptin, when the low, but not the high dose of TNF-alpha is employed. In conclusion, TNF-alpha, on a dose-dependent way, modulates insulin and leptin signaling and action in the hypothalamus.


Subject(s)
Cell Respiration/drug effects , Eating/drug effects , Hypothalamus/drug effects , Insulin/metabolism , Leptin/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Forkhead Transcription Factors/metabolism , Hypothalamus/metabolism , Immunoblotting , Immunoprecipitation , Insulin/administration & dosage , Insulin/pharmacology , Janus Kinase 2/metabolism , Leptin/administration & dosage , Leptin/pharmacology , Male , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Time Factors , Tumor Necrosis Factor-alpha/administration & dosage
19.
FEBS Lett ; 580(19): 4625-31, 2006 Aug 21.
Article in English | MEDLINE | ID: mdl-16876161

ABSTRACT

TNF-alpha acts on the hypothalamus modulating food intake and energy expenditure through mechanisms incompletely elucidated. Here, we explore the hypothesis that, to modulate insulin-induced anorexigenic signaling in hypothalamus, TNF-alpha requires the synthesis of NO. TNF-alpha activates signal transduction through JNK and p38 in hypothalamus, peaking at 10(-8) M. This is accompanied by the induction of expression of the inducible and neuronal forms of NOS, in both cases peaking at 10(-12) M. In addition, TNF-alpha stimulates NOS catalytic activity. Pre-treatment with TNF-alpha at a low dose (10(-12) M) inhibits insulin-dependent anorexigenic signaling, and this effect is abolished in iNOS but not in nNOS knockout mice.


Subject(s)
Feeding Behavior/physiology , Hypothalamus/drug effects , Insulin/physiology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Dose-Response Relationship, Drug , Hypothalamus/physiology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...