Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Nutr Biochem ; 119: 109372, 2023 09.
Article in English | MEDLINE | ID: mdl-37169229

ABSTRACT

High-fat diet consumption causes hypothalamic inflammation, dysregulating the leptin pathway, which, in turn, compromises the modulation of hypothalamic neuronal activities and predisposes obesity development. Intermittent fasting (IF) and exercise training (ET) have been demonstrated as efficient interventions to modulate hypothalamic inflammation and neuronal activity. However, no studies have evaluated whether combining these interventions could induce better results in reestablishing hypothalamic homeostasis disrupted by high-fat diet intake. The 8-week-old male C57BL/6 mice were randomly assigned into 2 groups: sedentary mice fed a standard diet (CT), and sedentary mice fed a high-fat diet (HF). After 8 weeks of an HF diet, part of the HF group (now 16 weeks old) was randomly subjected to different interventions for 6 weeks: HF-IF = HF diet mice submitted to IF; HF-T = HF diet mice submitted to ET; HF-IFT = HF diet mice submitted to IF and ET. All interventions decreased the body weight gain induced by high-fat diet intake, associated with reduced calorie consumption in week 14. Only the HF-IFT group presented improved serum insulin, leptin, resistin, and Tnf-alpha levels concomitantly with decreased hypothalamic inflammation. The HF-IFT group also demonstrated increased Pomc mRNA expression associated with enhanced pSTAT3 expression in the hypothalamic arcuate and ventromedial hypothalamic nuclei. Our data indicate that the beneficial effects of the combination of IF and ET on energy homeostasis are associated with increased leptin sensitivity in the hypothalamic arcuate nucleus and ventromedial hypothalamic nucleus, which is likely due to an improvement in hypothalamic inflammatory pathways in these nuclei.


Subject(s)
Diet, High-Fat , Leptin , Male , Mice , Animals , Diet, High-Fat/adverse effects , Intermittent Fasting , Dietary Fats/pharmacology , Mice, Inbred C57BL , Hypothalamus/metabolism , Inflammation/metabolism
2.
Nutrition ; 93: 111430, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34479044

ABSTRACT

OBJECTIVES: Based on taurine's beneficial roles in metabolic diseases in rodents and obese individuals, we investigated the effects of taurine supplementation on adipose tissue using transcriptome analysis, 3T3-L1 adipocytes, and subcutaneous white adipose tissue (scWAT) of obese women. METHODS: First, we applied bioinformatics analysis to evaluate the effect of the taurine synthesis pathway on the adipose tissue of several BXD mice strains. After that, using 3T3-L1 adipocytes, we investigated the effects of different taurine doses in proteins related to insulin signaling, lipid oxidation, and mitochondrial function. Finally, we evaluated the effects of taurine supplementation (3 grams, 8 wk) on the same proteins in the scWAT of obese women. RESULTS: The transcriptome analysis showed that the taurine biosynthesis pathway was positively associated with insulin signaling and mitochondrial metabolism in the scWAT of BXD mice. The experiments using 3T3-L1 cells highlighted that the taurine dosage has an essential function in taurine synthesis, insulin, and mitochondrial markers. In contrast, the 8-wk taurine administration did not change the basal insulin, proteins of the taurine synthesis or insulin pathways, lipid oxidation, or mitochondrial metabolism in the scWAT of obese women. CONCLUSIONS: For the first time, to our knowledge, we showed that supplementation with 3 g of taurine for 8 wk promoted no effect in the insulin signaling pathway in the scWAT of obese women. These findings bring new perspectives to investigate different taurine doses and the intervention period for human studies owing to the potential antiobesity activity of taurine.


Subject(s)
Insulin , Taurine , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Insulin/metabolism , Mice , Mitochondria , Obesity/drug therapy , Obesity/metabolism , Signal Transduction , Taurine/pharmacology
3.
Sci Rep ; 11(1): 3813, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33589652

ABSTRACT

The maintenance of mitochondrial activity in hypothalamic neurons is determinant to the control of energy homeostasis in mammals. Disturbs in the mitochondrial proteostasis can trigger the mitonuclear imbalance and mitochondrial unfolded protein response (UPRmt) to guarantee the mitochondrial integrity and function. However, the role of mitonuclear imbalance and UPRmt in hypothalamic cells are unclear. Combining the transcriptomic analyses from BXD mice database and in vivo experiments, we demonstrated that physical training alters the mitochondrial proteostasis in the hypothalamus of C57BL/6J mice. This physical training elicited the mitonuclear protein imbalance, increasing the mtCO-1/Atp5a ratio, which was accompanied by high levels of UPRmt markers in the hypothalamus. Also, physical training increased the maximum mitochondrial respiratory capacity in the brain. Interestingly, the transcriptomic analysis across several strains of the isogenic BXD mice revealed that hypothalamic mitochondrial DNA-encoded genes were negatively correlated with body weight and several genes related to the orexigenic response. As expected, physical training reduced body weight and food intake. Interestingly, we found an abundance of mt-CO1, a mitochondrial DNA-encoded protein, in NPY-producing neurons in the lateral hypothalamus nucleus of exercised mice. Collectively, our data demonstrated that physical training altered the mitochondrial proteostasis and induced the mitonuclear protein imbalance and UPRmt in hypothalamic cells.


Subject(s)
Energy Metabolism/genetics , Mitochondria/genetics , Proteostasis/genetics , Unfolded Protein Response/genetics , Animals , Humans , Hypothalamus/metabolism , Mice , Mitochondria/metabolism , Neurons/metabolism , Physical Conditioning, Animal/physiology
4.
J Anat ; 238(3): 743-750, 2021 03.
Article in English | MEDLINE | ID: mdl-33094520

ABSTRACT

The anatomy of the hypothalamus includes many nuclei and a complex network of neurocircuits. In this context, some hypothalamic nuclei reside closer to the blood-brain barrier, allowing communication with the peripheral organs through some molecules, such as leptin. Leptin is considered the main adipokine for energy homeostasis control. Furthermore, leptin signalling in the hypothalamus can communicate with insulin signalling through the activation of phosphoinositide 3-kinase (PI3k). Previous data suggest that isoforms of PI3k are necessary to mediate insulin action in the hypothalamus. However, obese animals show impairment in the central signalling of these hormones. Thus, in the current study, we evaluated the role of acute exercise in the leptin and insulin pathways in the hypothalamus, as well as in food intake control in obese mice. Although acute physical exercise was not able to modulate leptin signalling, this protocol suppressed the increase in the suppressor of cytokine signalling 3 (SOCS3) protein levels. In addition, acute exercise increased the content of PI3k-p110α protein in the hypothalamus. The exercised animals showed a strong tendency to reduction in cumulative food intake. For the first time, our results indicate physical exercise can increase PI3k-p110α protein content in the hypothalamus of obese mice and regulate food intake.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Obesity/therapy , Physical Conditioning, Animal/physiology , Animals , Male , Mice , Obesity/metabolism
5.
Nutrients ; 12(3)2020 Feb 28.
Article in English | MEDLINE | ID: mdl-32121154

ABSTRACT

The present study verified the responses of proteins related to the autophagy pathway after 10 h of fast with resistance exercise and protein ingestion in skeletal muscle and liver samples. The rats were distributed into five experimental groups: control (CT; sedentary and without gavage after fast), exercise immediately (EXE-imm; after fast, rats were submitted to the resistance protocol and received water by gavage immediately after exercise), exercise after 1 h (EXE-1h; after fast, rats were submitted to the resistance protocol and received water by gavage 1 h after exercise), exercise and supplementation immediately after exercise (EXE/Suppl-imm; after fast, rats were submitted to the resistance protocol and received a mix of casein: whey protein 1:1 (w/w) by gavage immediately after exercise), exercise and supplementation 1 h after exercise (EXE/Suppl-1h; after fast, rats were submitted to the resistance protocol and received a mix of casein: whey protein 1:1 (w/w) by gavage 1 h after exercise). In summary, the current findings show that the combination of fasting, acute resistance exercise, and protein blend ingestion (immediately or 1 h after the exercise stimulus) increased the serum levels of leucine, insulin, and glucose, as well as the autophagy protein contents in skeletal muscle, but decreased other proteins related to the autophagic pathway in the liver. These results deserve further mechanistic investigations since athletes are combining fasting with physical exercise to enhance health and performance outcomes.


Subject(s)
Autophagy , Biomarkers/metabolism , Dietary Proteins/administration & dosage , Fasting/physiology , Liver/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Resistance Training , Albumins/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Blood Glucose/metabolism , Body Weight/drug effects , Dietary Proteins/pharmacology , Eating , Fasting/blood , Gene Expression Regulation/drug effects , Insulin/blood , Leucine/blood , Liver/drug effects , Male , Muscle, Skeletal/drug effects , Organ Size/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Triglycerides/blood
6.
Eur J Nutr ; 59(6): 2427-2437, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31494696

ABSTRACT

PURPOSE: Nicotinamide riboside (NR) acts as a potent NAD+ precursor and improves mitochondrial oxidative capacity and mitochondrial biogenesis in several organisms. However, the effects of NR supplementation on aerobic performance remain unclear. Here, we evaluated the effects of NR supplementation on the muscle metabolism and aerobic capacity of sedentary and trained mice. METHODS: Male C57BL/6 J mice were supplemented with NR (400 mg/Kg/day) over 5 and 10 weeks. The training protocol consisted of 5 weeks of treadmill aerobic exercise, for 60 min a day, 5 days a week. Bioinformatic and physiological assays were combined with biochemical and molecular assays to evaluate the experimental groups. RESULTS: NR supplementation by itself did not change the aerobic performance, even though 5 weeks of NR supplementation increased NAD+ levels in the skeletal muscle. However, combining NR supplementation and aerobic training increased the aerobic performance compared to the trained group. This was accompanied by an increased protein content of NMNAT3, the rate-limiting enzyme for NAD + biosynthesis and mitochondrial proteins, including MTCO1 and ATP5a. Interestingly, the transcriptomic analysis using a large panel of isogenic strains of BXD mice confirmed that the Nmnat3 gene in the skeletal muscle is correlated with several mitochondrial markers and with different phenotypes related to physical exercise. Finally, NR supplementation during aerobic training markedly increased the amount of type I fibers in the skeletal muscle. CONCLUSION: Taken together, our results indicate that NR may be an interesting strategy to improve mitochondrial metabolism and aerobic capacity.


Subject(s)
Aerobiosis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Niacinamide/analogs & derivatives , Pyridinium Compounds/metabolism , Pyridinium Compounds/pharmacology , Animals , Cell Respiration/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Niacinamide/metabolism , Niacinamide/pharmacology
7.
J Cell Biochem ; 120(10): 18186-18192, 2019 10.
Article in English | MEDLINE | ID: mdl-31144370

ABSTRACT

Obesity and aging lead to abnormal transforming growth factor-ß1 (TGF-ß1) signaling in the hypothalamus, triggering the imbalance on glucose metabolism and energy homeostasis. Here, we determine the effect of acute exercise on TGF-ß1 expression in the hypothalamus of two models of obesity in mice. The bioinformatics analysis was performed to evaluate the correlation between hypothalamic Tgf-ß1 messenger RNA (mRNA) and genes related to thermogenesis in the brown adipose tissue (BAT) by using a large panel of isogenic BXD mice. Thereafter, leptin-deficient (ob/ob) mice and obese C57BL/6 mice fed on a high-fat diet (HFD) were submitted to the acute exercise protocol. Transcriptomic analysis by using BXD mouse reference population database revealed that hypothalamic Tgf-ß1 mRNA is negatively correlated with genes related to thermogenesis in brown adipose tissue of BXD mice, such as peroxisome proliferator-activated receptor gamma coactivator and is positively correlated with respiratory exchange ratio. In agreement with these results, leptin-deficient (ob/ob) and HFD-fed mice displayed high levels of Tgf-ß1 mRNA in the hypothalamus and reduction of Pgc1α mRNA in BAT. Interestingly, an acute exercise session reduced TGF-ß1 expression in the hypothalamus, increased Pgc1α mRNA in the BAT and reduced food consumption in obese mice. Our results demonstrated that acute physical exercise suppressed hypothalamic TGF-ß1 expression, increasing Pgc1α mRNA in BAT in obese mice.


Subject(s)
Down-Regulation , Hypothalamus/metabolism , Obesity/genetics , Physical Conditioning, Animal/physiology , Transforming Growth Factor beta1/genetics , Adipose Tissue, Brown/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Gene Expression Profiling/methods , Leptin/deficiency , Leptin/genetics , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thermogenesis/genetics , Transforming Growth Factor beta1/metabolism
8.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1126-1137, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30738810

ABSTRACT

In experimental obesity, the hypothalamus is affected by an inflammatory response activated by dietary saturated fats. This inflammation is triggered as early as one day after exposure to a high-fat diet, and during its progression, there is recruitment of inflammatory cells from the systemic circulation. The objective of the present study was identifying chemokines potentially involved in the development of hypothalamic diet-induced inflammation. In order to identify chemokines potentially involved in this process, we performed a real-time PCR array that determined Ackr2 as one of the transcripts undergoing differential regulation in obese-prone as compared to obese-resistant mice fed a high-fat diet for three days. ACKR2 is a decoy receptor that acts as an inhibitor of the signals generated by several CC inflammatory chemokines. Our results show that Ackr2 expression is rapidly induced after exposure to dietary fats both in obese-prone and obese-resistant mice. In immunofluorescence studies, ACKR2 was detected in hypothalamic neurons expressing POMC and NPY and also in microglia and astrocytes. The lentiviral overexpression of ACKR2 in the hypothalamus reduced diet-induced hypothalamic inflammation; however, there was no change in spontaneous caloric intake and body mass. Nevertheless, the overexpression of ACKR2 resulted in improvement of glucose tolerance, which was accompanied by reduced insulin secretion and increased whole body insulin sensitivity. Thus, ACKR2 is a decoy chemokine receptor expressed in most hypothalamic cells that is modulated by dietary intervention and acts to reduce diet-induced inflammation, leading to improved glucose tolerance due to improved insulin action.


Subject(s)
Gene Expression Profiling , Glucose/metabolism , Hypothalamus/metabolism , Inflammation/genetics , Obesity/genetics , Receptors, Chemokine/genetics , Animals , Astrocytes/metabolism , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat/adverse effects , Glucose Tolerance Test , Hypothalamus/cytology , Inflammation/etiology , Inflammation/metabolism , Insulin Resistance/genetics , Male , Mice , Neurons/metabolism , Obesity/etiology , Obesity/metabolism , Receptors, Chemokine/metabolism
9.
Brain Behav Immun ; 78: 78-90, 2019 05.
Article in English | MEDLINE | ID: mdl-30660601

ABSTRACT

Obesity-associated hypothalamic inflammation plays an important role in the development of defective neuronal control of whole body energy balance. Because dietary fats are the main triggers of hypothalamic inflammation, we hypothesized that CD1, a lipid-presenting protein, may be involved in the hypothalamic inflammatory response in obesity. Here, we show that early after the introduction of a high-fat diet, CD1 expressing cells gradually appear in the mediobasal hypothalamus. The inhibition of hypothalamic CD1 reduces diet-induced hypothalamic inflammation and rescues the obese and glucose-intolerance phenotype of mice fed a high-fat diet. Conversely, the chemical activation of hypothalamic CD1 further increases diet-induced obesity and hypothalamic inflammation. A bioinformatics analysis revealed that hypothalamic CD1 correlates with transcripts encoding for proteins known to be involved in diet-induced hypothalamic abnormalities in obesity. Thus, CD1 is involved in at least part of the hypothalamic inflammatory response in diet-induced obesity and its modulation affects the body mass phenotype of mice.


Subject(s)
Antigens, CD1/metabolism , Hypothalamus/immunology , Obesity/metabolism , Animals , Antigens, CD1/immunology , Computational Biology/methods , Diet, High-Fat , Dietary Fats , Energy Metabolism , Inflammation/metabolism , Lymphocytes/metabolism , Male , Mice , Obesity/immunology
10.
Life Sci ; 211: 1-7, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30195617

ABSTRACT

AIMS: Nicotinamide Riboside (NR) is a NAD+ booster with wide physiological repercussion including the improvement on glucose and lipid homeostasis, increasing the life expectancy in mammals. However, the effects of NR on metabolism are only partially known. Here, we evaluated the effects of NR on the thermogenic response, highlighting the brown adipose tissue (BAT) in lean mice. MAIN METHODS: Male C57BL/67 mice were supplement with NR (400 mg/Kg/day) during 5 weeks. The Comprehensive Lab Animal Monitoring System (CLAMS) and thermographic images were used to evaluated the physiological effects of NR treatment. The BAT were extracted and analyzed by Western Blotting and qPCR. Also, bioinformatics analyses were performed to establish the connection between the NAD+ synthesis pathway in BAT and thermogenic response in several isogenic strains of BXD mice. KEY FINDINGS: Transcriptomic analysis revealed that genes involved in NAD+ synthesis (Nampt and Nmnat1) in the BAT were negatively correlated with body weight and fat mass. The heat map showed a strong positive correlation between Nampt and Ucp1 mRNA in BAT and body temperature in several strains of BXD lean mice. The experimental approaches demonstrated that oral NR supplementation reduced the abdominal visceral fat depots, with discrete impact on oxygen consumption in C57BL/6J mice. Interestingly, NR significantly increased the body temperature, and this phenomenon was accompanied by high levels of UCP1 protein content and Pgc1α mRNA in BAT. SIGNIFICANCE: This study demonstrated the oral NR supplementation was sufficient to induce the thermogenic response in lean mice changing the BAT metabolism.


Subject(s)
Adipose Tissue, Brown/physiology , Body Temperature/drug effects , Gene Expression Regulation/drug effects , Niacinamide/analogs & derivatives , Thermogenesis/drug effects , Thinness/drug therapy , Adipose Tissue, Brown/drug effects , Administration, Oral , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Niacinamide/administration & dosage , Niacinamide/pharmacology , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Pyridinium Compounds , Thinness/metabolism , Thinness/pathology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
11.
J Cell Physiol ; 233(12): 9426-9436, 2018 12.
Article in English | MEDLINE | ID: mdl-30063084

ABSTRACT

Hypothalamic sphingosine-1-phosphate receptor 1 (S1PR1), the G protein-coupled receptor 1 of sphingosine-1-phosphate, has been described as a modulator in the control of energy homeostasis in rodents. However, this mechanism is still unclear. Here, we evaluate the role of interleukin 6 (IL-6) associated with acute physical exercise in the control of the hypothalamic S1PR1-signal transducer and activator of transcription 3 (STAT3) axis. Acute exercise session and an intracerebroventricular IL-6 injection increased S1PR1 protein content and STAT3 phosphorylation in the hypothalamus of lean and obese mice accompanied by a reduction in food consumption. Transcriptome analysis indicated a strong positive correlation between Il-6 and S1pr1 messenger RNA in several tissues of genetically diverse BXD mice strains and humans, including in the hypothalamus. Interestingly, exercise failed to stimulate the S1PR1-STAT3 axis in IL-6 knockout mice and the disruption of hypothalamic-specific IL-6 action blocked the anorexigenic effects of exercise. Taken together, our results indicate that physical exercise modulates the S1PR1 protein content in the hypothalamus, through the central action of IL-6.


Subject(s)
Hypothalamus/metabolism , Interleukin-6/metabolism , Physical Conditioning, Animal , Receptors, Lysosphingolipid/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Humans , Injections, Intraventricular , Interleukin-6/administration & dosage , Interleukin-6/genetics , Male , Mice, Inbred C57BL , Mice, Obese , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/genetics , Sphingosine-1-Phosphate Receptors
12.
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
13.
Brain Behav Immun ; 73: 550-561, 2018 10.
Article in English | MEDLINE | ID: mdl-29935943

ABSTRACT

Hypothalamic hypoxia-inducible factor-1 (HIF-1) can regulate whole-body energy homeostasis in response to changes in blood glucose, suggesting that it acts as a sensor for systemic energy stores. Here, we hypothesized that hypothalamic HIF-1 could be affected by diet-induced obesity (DIO). We used eight-week old, male C57Bl6 mice, fed normal chow diet or with high fat diet for 1, 3, 7, 14 and 28 days. The expression of HIF-1alpha and HIF-1beta was measured by PCR and western blotting and its hypothalamic distribution was evaluated by fluorescence microscopy. Inhibition of HIF-1beta in arcuate nucleus of hypothalamus was performed using stereotaxic injection of shRNA lentiviral particles and animals were grouped under normal chow diet or high fat diet for 14 days. Using bioinformatics, we show that in humans, the levels of HIF-1 transcripts are directly correlated with those of hypothalamic transcripts for proteins involved in inflammation, regulation of apoptosis, autophagy, and the ubiquitin/proteasome system; furthermore, in rodents, hypothalamic HIF-1 expression is directly correlated with the phenotype of increased energy expenditure. In mice, DIO was accompanied by increased HIF-1 expression. The inhibition of hypothalamic HIF-1 by injection of an shRNA resulted in a further increase in body mass, a decreased basal metabolic rate, increased hypothalamic inflammation, and glucose intolerance. Thus, hypothalamic HIF-1 is increased during DIO, and its inhibition worsens the obesity-associated metabolic phenotype. Thus, hypothalamic HIF-1 emerges as a target for therapeutic intervention against obesity.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Obesity/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Blood Glucose/metabolism , Body Weight , Diet, High-Fat/adverse effects , Down-Regulation , Energy Metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/physiopathology
14.
Cytokine ; 103: 69-76, 2018 03.
Article in English | MEDLINE | ID: mdl-29331586

ABSTRACT

The association between excessive training sessions (i.e., overtraining/OT) and periods of inadequate recovery is linked to the nonfunctional overreaching (NFOR) state, which is defined as an unexplained decrement or stagnation of performance. The cytokine hypothesis of OT considers that pro-inflammatory cytokines are responsible by the NFOR state-induced performance decrement. Investigations using rodent models of OT verified increased levels of pro-inflammatory cytokines in hypothalamus, liver, serum and skeletal muscle samples. Recently, our research group observed that a 2-week total recovery period was not able to re-establish the NFOR state-induced performance decrement. As the responses of anti- and pro-inflammatory cytokines were not measured, we aimed to investigate the effects of 2-week total recovery period on the protein contents of IL-1beta, IL-6, IL-10, IL-15, TNF-alpha and SOCS-3 in serum and skeletal muscle samples of overtrained mice. Also, a bioinformatics analysis was performed to investigate the correlations of IL-1beta, IL-6, IL-10, IL-15, TNF-alpha and SOCS-3 in skeletal muscle with locomotor activity. In summary, the 2-week total recovery period upregulated the anti-inflammatory cytokines and normalized the pro-inflammatory cytokines without a concomitant re-establishment of performance.


Subject(s)
Cytokines/metabolism , Hypothalamus/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Animals , Male , Mice
15.
Sci Transl Med ; 8(361): 361ra139, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27798264

ABSTRACT

Neuromuscular diseases are often caused by inherited mutations that lead to progressive skeletal muscle weakness and degeneration. In diverse populations of normal healthy mice, we observed correlations between the abundance of mRNA transcripts related to mitochondrial biogenesis, the dystrophin-sarcoglycan complex, and nicotinamide adenine dinucleotide (NAD+) synthesis, consistent with a potential role for the essential cofactor NAD+ in protecting muscle from metabolic and structural degeneration. Furthermore, the skeletal muscle transcriptomes of patients with Duchene's muscular dystrophy (DMD) and other muscle diseases were enriched for various poly[adenosine 5'-diphosphate (ADP)-ribose] polymerases (PARPs) and for nicotinamide N-methyltransferase (NNMT), enzymes that are major consumers of NAD+ and are involved in pleiotropic events, including inflammation. In the mdx mouse model of DMD, we observed significant reductions in muscle NAD+ levels, concurrent increases in PARP activity, and reduced expression of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme for NAD+ biosynthesis. Replenishing NAD+ stores with dietary nicotinamide riboside supplementation improved muscle function and heart pathology in mdx and mdx/Utr-/- mice and reversed pathology in Caenorhabditis elegans models of DMD. The effects of NAD+ repletion in mdx mice relied on the improvement in mitochondrial function and structural protein expression (α-dystrobrevin and δ-sarcoglycan) and on the reductions in general poly(ADP)-ribosylation, inflammation, and fibrosis. In combination, these studies suggest that the replenishment of NAD+ may benefit patients with muscular dystrophies or other neuromuscular degenerative conditions characterized by the PARP/NNMT gene expression signatures.


Subject(s)
Muscle, Skeletal/physiopathology , Muscular Dystrophies/pathology , NAD/chemistry , Poly ADP Ribosylation , Adenosine Diphosphate/chemistry , Animals , Caenorhabditis elegans , Cell Line , Cytokines/chemistry , Fibrosis/pathology , Gene Expression Profiling , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscular Diseases/pathology , Nicotinamide Phosphoribosyltransferase/chemistry , Nitrosamines/chemistry , RNA, Messenger/metabolism , Tyramine/analogs & derivatives , Tyramine/chemistry
16.
Nat Commun ; 5: 4859, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25255053

ABSTRACT

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.


Subject(s)
Energy Metabolism , Hypothalamus/metabolism , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Homeostasis , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Rats , Rats, Wistar , Receptors, Lysosphingolipid/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors
17.
Am J Physiol Endocrinol Metab ; 305(5): E649-59, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23880311

ABSTRACT

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 (IRß 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.


Subject(s)
Hypothalamus/metabolism , Inflammation/metabolism , Insulin/metabolism , Leptin/metabolism , Obesity/metabolism , Physical Conditioning, Animal/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Blotting, Western , Corticosterone/urine , Hypothalamus/enzymology , Immunohistochemistry , Inflammation/enzymology , Insulin/blood , Interleukin-6/blood , Interleukin-6/metabolism , Leptin/blood , Male , Mice , Mice, Obese , Obesity/enzymology , Random Allocation , Rats , Rats, Wistar , Signal Transduction , Specific Pathogen-Free Organisms
18.
Diabetes ; 62(1): 137-48, 2013 01.
Article in English | MEDLINE | ID: mdl-22966070

ABSTRACT

Mutation of tub gene in mice induces obesity, suggesting that tub could be an important regulator of energy balance. In the current study, we investigated whether insulin, leptin, and obesity can modulate Tub in vivo in hypothalamic nuclei, and we investigated possible consequences on energy balance, neuropeptide expression, and hepatic glucose metabolism. Food intake, metabolic characteristics, signaling proteins, and neuropeptide expression were measured in response to fasting and refeeding, intracerebroventricular insulin and leptin, and Tub antisense oligonucleotide (ASO). Tub tyrosine phosphorylation (Tub-p-tyr) is modulated by nutritional status. Tub is a substrate of insulin receptor tyrosine kinase (IRTK) and leptin receptor (LEPR)-Janus kinase 2 (JAK2) in hypothalamic nuclei. After leptin or insulin stimulation, Tub translocates to the nucleus. Inhibition of Tub expression in hypothalamus by ASO increased food intake, fasting blood glucose, and hepatic glucose output, decreased O(2) consumption, and blunted the effect of insulin or leptin on proopiomelanocortin, thyroid-releasing hormone, melanin-concentrating hormone, and orexin expression. In hypothalamus of mice administered a high-fat diet, there is a reduction in leptin and insulin-induced Tub-p-tyr and nuclear translocation, which is reversed by reducing protein tyrosine phosphatase 1B expression. These results indicate that Tub has a key role in the control of insulin and leptin effects on food intake, and the modulation of Tub may contribute to insulin and leptin resistance in DIO mice.


Subject(s)
Hypothalamus/physiology , Insulin/pharmacology , Leptin/pharmacology , Proteins/physiology , Signal Transduction/physiology , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing , Animals , Fasting , Janus Kinase 2/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/pharmacology , Phospholipase C beta/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Proteins/antagonists & inhibitors
19.
Metabolism ; 62(2): 171-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22898253

ABSTRACT

Hypothalamic 5'-adenosine monophosphate-activated protein kinase (AMPK) senses intracellular metabolic stress, i.e., an increase in the cellular AMP:ATP ratio, and integrates diverse hormonal and nutritional signals to restore energy balance. Recent evidence suggests that different nutrients can modulate AMPK activity in the hypothalamus, thereby controlling weight gain through a leptin-independent mechanism. Understanding the mechanisms by which nutrients control hypothalamic AMPK activity is crucial to the development of effective nutritional interventions for the treatment of food intake-related disorders, such as anorexia and obesity. This article highlights the current evidence for the intricate relationship between nutrients and hypothalamic AMPK activity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Eating/physiology , Hypothalamus/metabolism , Animals , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Hypothalamus/enzymology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL