Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Chem Biol Interact ; 256: 178-87, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27417254

ABSTRACT

High-fat (HF) diet-induced obesity is associated with hypothalamic leptin resistance and low grade chronic inflammation, which largely impairs the neuroregulation of negative energy balance. Neuroregulation of negative energy balance is largely controlled by the mediobasal and paraventricular nuclei regions of the hypothalamus via leptin signal transduction. Recently, a derivative of oleanolic acid, bardoxolone methyl (BM), has been shown to have anti-inflammatory effects. We tested the hypothesis that BM would prevent HF diet-induced obesity, hypothalamic leptin resistance, and inflammation in mice fed a HF diet. Oral administration of BM via drinking water (10 mg/kg daily) for 21 weeks significantly prevented an increase in body weight, energy intake, hyperleptinemia, and peripheral fat accumulation in mice fed a HF diet. Furthermore, BM treatment prevented HF diet-induced decreases in the anorexigenic effects of peripheral leptin administration. In the mediobasal and paraventricular nuclei regions of the hypothalamus, BM administration prevented HF diet-induced impairments of the downstream protein kinase b (Akt) pathway of hypothalamic leptin signalling. BM treatment also prevented an increase in inflammatory cytokines, tumour necrosis factor alpha (TNFα) and interleukin 6 (IL-6) in these two hypothalamic regions. These results identify a potential novel neuropharmacological application for BM in preventing HF diet-induced obesity, hypothalamic leptin resistance, and inflammation.


Subject(s)
Anti-Obesity Agents/therapeutic use , Hypothalamus/drug effects , Leptin/metabolism , Obesity/prevention & control , Oleanolic Acid/analogs & derivatives , Signal Transduction/drug effects , Animals , Body Weight/drug effects , Diet, High-Fat/adverse effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Hypothalamus/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/physiopathology , Inflammation/prevention & control , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Oleanolic Acid/therapeutic use
2.
J Histochem Cytochem ; 64(4): 237-55, 2016 04.
Article in English | MEDLINE | ID: mdl-26920068

ABSTRACT

Obesity induces chronic, low-grade inflammation, which increases the risk of colon cancer. We investigated the preventive effects of Bardoxolone methyl (BARD) on high-fat diet (HFD)-induced inflammation in a mouse colon. Male C57BL/6J mice (n=7) were fed a HFD (HFD group), HFD plus BARD (10 mg/kg) in drinking water (HFD/BARD group), or normal laboratory chow diet (LFD group) for 21 weeks. In HFD mice, BARD reduced colon thickness and decreased colon weight per length. This was associated with an increase in colon crypt depth and the number of goblet cells per crypt. BARD reduced the expression of F4/80 and CD11c but increased CD206 and IL-10, indicating an anti-inflammatory effect. BARD prevented an increase of the intracellular pro-inflammatory biomarkers (NF-қB, p NF-қB, IL-6, TNF-α) and cell proliferation markers (Cox2 and Ki67). BARD prevented fat deposition in the colon wall and prevented microbial population changes. Overall, we report the preventive effects of BARD on colon inflammation in HFD-fed mice through its regulation of macrophages, NF-қB, cytokines, Cox2 and Ki67, fat deposition and microflora.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colon/drug effects , Diet, High-Fat/adverse effects , Inflammation/etiology , Inflammation/prevention & control , Oleanolic Acid/analogs & derivatives , Animals , Colon/immunology , Colon/pathology , Cytokines/immunology , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/immunology , Male , Mice , Mice, Inbred C57BL , Oleanolic Acid/therapeutic use
3.
Chem Biol Interact ; 243: 10-8, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26612656

ABSTRACT

Obesity caused by the consumption of a high-fat (HF) diet is a major risk factor for the development of associated complications, such as heart and kidney failure. A semi-synthetic triterpenoid, bardoxolone methyl (BM) was administrated to mice fed a HF diet for 21 weeks to determine if it would prevent the development of obesity-associated cardiac and renal pathophysiologies. Twelve week old male C57BL/6J mice were fed a lab chow (LC), HF (40% fat), or a HF diet supplemented with 10 mg/kg/day BM in drinking water. After 21 weeks, the left ventricles of hearts and cortex of kidneys of mice were collected for analysis. Histological analysis revealed that BM prevented HF diet-induced development of structural changes in the heart and kidneys. BM prevented HF diet-induced decreases in myocyte number in cardiac tissue, although this treatment also elevated cardiac endothelin signalling molecules. In the kidneys, BM administration prevented HF diet-induced renal corpuscle hypertrophy and attenuated endothelin signalling. Furthermore, in both the hearts and kidneys of mice fed a HF diet, BM administration prevented HF diet-induced increases in fat accumulation, macrophage infiltration and tumour necrosis factor alpha (TNFα) gene expression. These findings suggest that BM prevents HF diet-induced developments of cardiac and renal pathophysiologies in mice fed a chronic HF diet by preventing inflammation. Moreover, these results suggest that BM has the potential as a therapeutic for preventing obesity-induced cardiac and renal pathophysiologies.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diet, High-Fat/adverse effects , Heart/drug effects , Hypertrophy/prevention & control , Inflammation/prevention & control , Kidney/drug effects , Oleanolic Acid/analogs & derivatives , Animals , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Fats/analysis , Heart/physiopathology , Hypertrophy/complications , Hypertrophy/pathology , Hypertrophy/physiopathology , Inflammation/complications , Inflammation/pathology , Inflammation/physiopathology , Kidney/pathology , Kidney/physiopathology , Male , Mice, Inbred C57BL , Myocardium/pathology , Oleanolic Acid/therapeutic use
4.
ScientificWorldJournal ; 2015: 549352, 2015.
Article in English | MEDLINE | ID: mdl-26618193

ABSTRACT

Mesenteric fat belongs to visceral fat. An increased deposition of mesenteric fat contributes to obesity associated complications such as type 2 diabetes and cardiovascular diseases. We have investigated the therapeutic effects of bardoxolone methyl (BARD) on mesenteric adipose tissue of mice fed a high-fat diet (HFD). Male C57BL/6J mice were administered oral BARD during HFD feeding (HFD/BARD), only fed a high-fat diet (HFD), or fed low-fat diet (LFD) for 21 weeks. Histology and immunohistochemistry were used to analyse mesenteric morphology and macrophages, while Western blot was used to assess the expression of inflammatory, oxidative stress, and energy expenditure proteins. Supplementation of drinking water with BARD prevented mesenteric fat deposition, as determined by a reduction in large adipocytes. BARD prevented inflammation as there were fewer inflammatory macrophages and reduced proinflammatory cytokines (interleukin-1 beta and tumour necrosis factor alpha). BARD reduced the activation of extracellular signal-regulated kinase (ERK) and Akt, suggesting an antioxidative stress effect. BARD upregulates energy expenditure proteins, judged by the increased activity of tyrosine hydroxylase (TH) and AMP-activated protein kinase (AMPK) and increased peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and uncoupling protein 2 (UCP2) proteins. Overall, BARD induces preventive effect in HFD mice through regulation of mesenteric adipose tissue.


Subject(s)
Diet, High-Fat/adverse effects , Intra-Abdominal Fat/drug effects , Obesity/prevention & control , Oleanolic Acid/analogs & derivatives , AMP-Activated Protein Kinases/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Energy Metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation/drug therapy , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Ion Channels/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Obesity/etiology , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Oxidative Stress , PPAR gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Uncoupling Protein 2
5.
Mol Nutr Food Res ; 59(12): 2371-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26314570

ABSTRACT

SCOPE: Obesity impairs cognition, and the leptin-induced increase of brain-derived neurotrophic factor (BDNF) and neurogenesis. Tea consumption improves cognition and increases brain activation in the prefrontal cortex. METHODS AND RESULTS: This study examined whether teasaponin, an active ingredient in tea, could improve memory and central leptin effects on neurogenesis in the prefrontal cortex of obese mice, and in vitro in cultured prefrontal cortical neurons. Teasaponin (10 mg/kg, intraperitoneal) for 21 days improved downstream leptin signaling (JAK2 and STAT3), and leptin's effect on BDNF, in the prefrontal cortex of high-fat diet (HFD) fed mice. Prefrontal cortical neurons were cultured with teasaponin and palmitic acid (the most abundant dietary saturated fatty acid) to examine their effects on neurogenesis and BDNF expression in response to leptin. Palmitic acid decreased leptin's effect on neurite outgrowth, postsynaptic density protein 95, and BDNF expression in cultured cortical neurons, which was reversed by teasaponin. CONCLUSION: Teasaponin improved the leptin sensitivity of prefrontal cortical neurons in obese mice or when treated by palmitic acid. This in turn increased BDNF expression and neurite growth. Therefore, teasaponin supplementation may be used to prevent obesity-associated neurodegeneration and improve cognitive function.


Subject(s)
Leptin/metabolism , Obesity/drug therapy , Prefrontal Cortex/drug effects , Saponins/pharmacology , Tea/chemistry , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Diet, High-Fat/adverse effects , Hyperinsulinism/drug therapy , Hyperinsulinism/metabolism , Insulin/blood , Leptin/blood , Leptin/pharmacology , Male , Mice, Inbred C57BL , Mice, Obese , Neurons/drug effects , Neurons/metabolism , Obesity/etiology , Obesity/physiopathology , Palmitic Acid/pharmacology , Prefrontal Cortex/physiopathology , STAT3 Transcription Factor/metabolism , Weight Gain/drug effects
6.
Nutrients ; 7(6): 4705-23, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26066016

ABSTRACT

Obesity results in changes in brown adipose tissue (BAT) morphology, leading to fat deposition, inflammation, and alterations in sympathetic nerve activity. Bardoxolone methyl (BARD) has been extensively studied for the treatment of chronic diseases. We present for the first time the effects of oral BARD treatment on BAT morphology and associated changes in the brainstem. Three groups (n = 7) of C57BL/6J mice were fed either a high-fat diet (HFD), a high-fat diet supplemented with BARD (HFD/BARD), or a low-fat diet (LFD) for 21 weeks. BARD was administered daily in drinking water. Interscapular BAT, and ventrolateral medulla (VLM) and dorsal vagal complex (DVC) in the brainstem, were collected for analysis by histology, immunohistochemistry and Western blot. BARD prevented fat deposition in BAT, demonstrated by the decreased accumulation of lipid droplets. When administered BARD, HFD mice had lower numbers of F4/80 and CD11c macrophages in the BAT with an increased proportion of CD206 macrophages, suggesting an anti-inflammatory effect. BARD increased phosphorylation of tyrosine hydroxylase in BAT and VLM. In the VLM, BARD increased energy expenditure proteins, including beta 3-adrenergic receptor (ß3-AR) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Overall, oral BARD prevented fat deposition and inflammation in BAT, and stimulated sympathetic nerve activity.


Subject(s)
Adipose Tissue, Brown/drug effects , Anti-Inflammatory Agents/pharmacology , Diet, High-Fat/adverse effects , Inflammation/prevention & control , Oleanolic Acid/analogs & derivatives , Adipose Tissue, Brown/metabolism , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Diet, Fat-Restricted , Energy Metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Oleanolic Acid/pharmacology , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism
7.
Mol Cell Endocrinol ; 412: 12-8, 2015 Sep 05.
Article in English | MEDLINE | ID: mdl-25986657

ABSTRACT

Epidemiological evidence suggests that the consumption of a diet high in n-6 polyunsaturated fatty acids (PUFA) is associated with the development of leptin resistance and obesity. We aim to examine the central effect of n-6 PUFA, arachidonic acid (ARA) on leptin sensitivity and leptin-regulated hepatic glucose and lipid metabolism. We found that intracerebroventricular injection of ARA (25 nmol/day) for 2.5 days reversed the effect of central leptin on hypothalamic JAK2, pSTAT3, pAkt, and pFOXO1 protein levels, which was concomitant with a pro-inflammatory response in the hypothalamus. ARA also attenuated the effect of central leptin on hepatic glucose and lipid metabolism by reversing the mRNA expression of the genes involved in gluconeogenesis (G6Pase, PEPCK), glucose transportation (GLUT2), lipogenesis (FAS, SCD1), and cholesterol synthesis (HMG-CoA reductase). These results indicate that an increased exposure to central n-6 PUFA induces central cellular leptin resistance with concomitant defective JAK2-STAT3 and PI3K-Akt signaling.


Subject(s)
Arachidonic Acid/pharmacology , Energy Metabolism , Leptin/physiology , Liver/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Animals , Energy Intake , Glucose/metabolism , Homeostasis , Hypothalamus/immunology , Hypothalamus/metabolism , Lipid Metabolism , Male , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus/immunology , Signal Transduction , Tyrosine 3-Monooxygenase/metabolism
8.
Mol Cell Endocrinol ; 412: 36-43, 2015 Sep 05.
Article in English | MEDLINE | ID: mdl-26001833

ABSTRACT

High-fat (HF) diet-induced obesity is a major risk factor for the development of insulin resistance and hepatic steatosis. We examined the hypothesis that bardoxolone methyl (BM) would prevent the development of insulin resistance and hepatic steatosis in mice fed a HF diet. C57BL/6J male mice were fed a lab chow (LC), HF (40% fat), or HF diet supplemented with 10 mg/kg/day BM orally for 21 weeks. Glucose metabolism was assessed using a glucose tolerance test (GTT) and insulin sensitivity test (IST). Signalling molecules involved in insulin resistance, inflammation, and lipid metabolism were examined in liver tissue via western blotting and RT-PCR. BM prevented HF diet-induced insulin resistance and alterations in the protein levels of protein tyrosine phosphatase 1B (PTP1B), forkhead box protein O1 (FOXO1) and BDNF, and expression of the insulin receptor (IR), IRS-1 and glucose-6-phosphatase (G6Pase) genes. Furthermore, BM prevented fat accumulation in the liver and decreases in the ß-oxidation gene, peroxisomal acyl-coenzyme A oxidase 1 (ACOX) in mice fed a HF diet. In the livers of HF fed mice, BM administration prevented HF diet-induced macrophage infiltration, inflammation as indicated by reduced IL-6 and signal transducer and activator of transcription 3 (STAT3) protein levels and TNFα mRNA expression, and increased nuclear factor-like 2 (Nrf2) mRNA expression and nuclear protein levels. These findings suggest that BM prevents HF diet induced insulin resistance and the development of hepatic steatosis in mice fed a chronic HF diet through modulation of molecules involved in insulin signalling, lipid metabolism and inflammation in the liver.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/prevention & control , Insulin Resistance , Lipid Metabolism/drug effects , Oleanolic Acid/analogs & derivatives , Animals , Cell Movement , Fatty Liver/blood , Fatty Liver/etiology , Insulin/blood , Liver/drug effects , Liver/immunology , Liver/metabolism , Liver/pathology , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , Obesity/blood , Obesity/etiology , Oleanolic Acid/pharmacology
9.
J Nutr Biochem ; 26(5): 541-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25724108

ABSTRACT

The consumption of diets rich in saturated fat largely contributes to the development of obesity in modern societies. A diet high in saturated fats can induce inflammation and impair leptin signaling in the hypothalamus. However, the role of saturated fatty acids on hypothalamic leptin signaling, and hepatic glucose and lipid metabolism remains largely undiscovered. In this study, we investigated the effects of intracerebroventricular (icv) administration of a saturated fatty acid, palmitic acid (PA, C16:0), on central leptin sensitivity, hypothalamic leptin signaling, inflammatory molecules and hepatic energy metabolism in C57BL/6J male mice. We found that the icv administration of PA led to central leptin resistance, evidenced by the inhibition of central leptin's suppression of food intake. Central leptin resistance was concomitant with impaired hypothalamic leptin signaling (JAK2-STAT3, PKB/Akt-FOXO1) and a pro-inflammatory response (TNF-α, IL1-ß, IL-6 and pIκBa) in the mediobasal hypothalamus and paraventricular hypothalamic nuclei. Furthermore, the pre-administration of icv PA blunted the effect of leptin-induced decreases in mRNA expression related to gluconeogenesis (G6Pase and PEPCK), glucose transportation (GLUT2) and lipogenesis (FAS and SCD1) in the liver of mice. Therefore, elevated central PA concentrations can induce pro-inflammatory responses and leptin resistance, which are associated with disorders of energy homeostasis in the liver as a result of diet-induced obesity.


Subject(s)
Glucose/metabolism , Leptin/pharmacology , Lipid Metabolism/drug effects , Liver/drug effects , Palmitic Acid/pharmacology , Animals , Hypothalamus/drug effects , Hypothalamus/enzymology , Hypothalamus/pathology , Inflammation/chemically induced , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Tyrosine 3-Monooxygenase/metabolism
10.
Chem Biol Interact ; 229: 1-8, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25637688

ABSTRACT

Key features of diet-induced obesity are visceral fat deposition, macrophage infiltration and inflammation that can lead to metabolic disorders. This study examined the effects of bardoxolone methyl (BARD) in preventing obesity and inflammation in the visceral fat of mice fed high-fat diet. Male C57BL/6J mice were fed a high-fat diet (HFD), a low-fat diet (LFD, i.e., lab chow diet) or a high-fat diet supplemented with BARD (HFD/BARD) for 21weeks. BARD at a dosage of 10mg/kg body weight was administered orally in drinking water. Histology, immunohistochemistry and Western blot were used for the analysis of epididymal adipose tissue. Morphological results demonstrated that HFD fed mice treated with BARD had smaller adipocytes and fewer macrophages present in epididymal adipose tissue than the HFD group. Furthermore, BARD administration reduced the inflammatory profile in this tissue by increasing the expression of nuclear factor of kappa-light-polypeptide gene enhancer in B-cells inhibitor, alpha (IκB-α) protein and decreasing the protein expression of tumour necrosis factor alpha (TNF-α). BARD also prevented oxidative stress reflected by a reduction in stress activated proteins, including signal transducer and activator of transcription 3 (STAT3), protein kinase B (Akt), extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK). BARD administration activated the sympathetic nervous system in epididymal adipose tissue assessed by the increased synthesis of tyrosine hydroxylase (TH) and uncoupling protein 2 (UCP2). The expression of inflammatory and sympathetic nervous system proteins in BARD mice fed a HFD was equivalent to that of the LFD control mice, indicating the anti-inflammatory and anti-obesity properties of this drug. In conclusion, the oral administration of BARD in HFD mice prevented fat deposition, inflammation and oxidative stress, and improved sympathetic activity in visceral fat. This study suggests a potential therapeutic role of BARD in preventing the development of obesity.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Diet, High-Fat/adverse effects , Inflammation/prevention & control , Intra-Abdominal Fat/drug effects , Obesity/prevention & control , Oleanolic Acid/analogs & derivatives , Adipocytes/cytology , Adipocytes/drug effects , Animals , Inflammation/immunology , Inflammation/metabolism , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/metabolism , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/immunology , Obesity/metabolism , Oleanolic Acid/therapeutic use , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/immunology
11.
Article in English | MEDLINE | ID: mdl-25584778

ABSTRACT

High fat (HF) diets are known to induce changes in synaptic plasticity in the forebrain leading to learning and memory impairments. Previous studies of oleanolic acid derivatives have found that these compounds can cross the blood-brain barrier to prevent neuronal cell death. We examined the hypothesis that the oleanolic acid derivative, bardoxolone methyl (BM) would prevent diet-induced cognitive deficits in mice fed a HF diet. C57BL/6J male mice were fed a lab chow (LC) (5% of energy as fat), a HF (40% of energy as fat), or a HF diet supplemented with 10mg/kg/day BM orally for 21weeks. Recognition memory was assessed by performing a novel object recognition test on the treated mice. Downstream brain-derived neurotrophic factor (BDNF) signalling molecules were examined in the prefrontal cortex (PFC) and hippocampus of mice via Western blotting and N-methyl-d-aspartate (NMDA) receptor binding. BM treatment prevented HF diet-induced impairment in recognition memory (p<0.001). In HF diet fed mice, BM administration attenuated alterations in the NMDA receptor binding density in the PFC (p<0.05), however, no changes were seen in the hippocampus (p>0.05). In the PFC and hippocampus of the HF diet fed mice, BM administration improved downstream BDNF signalling as indicated by increased protein levels of BDNF, phosphorylated tropomyosin related kinase B (pTrkB) and phosphorylated protein kinase B (pAkt), and increased phosphorylated AMP-activated protein kinase (pAMPK) (p<0.05). BM administration also prevented the HF diet-induced increase in the protein levels of inflammatory molecules, phosphorylated c-Jun N-terminal kinase (pJNK) in the PFC, and protein tyrosine phosphatase 1B (PTP1B) in both the PFC and hippocampus. In summary, these findings suggest that BM prevents HF diet-induced impairments in recognition memory by improving downstream BDNF signal transduction, increasing pAMPK, and reducing inflammation in the PFC and hippocampus.


Subject(s)
Diet, High-Fat/adverse effects , Memory Disorders , Oleanolic Acid/analogs & derivatives , Prefrontal Cortex/drug effects , Recognition, Psychology/drug effects , Signal Transduction/drug effects , Animals , Autoradiography , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Drug Administration Schedule , Exploratory Behavior/drug effects , Male , Memory Disorders/etiology , Memory Disorders/pathology , Memory Disorders/prevention & control , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , Oleanolic Acid/therapeutic use , Prefrontal Cortex/metabolism , Protein Binding/drug effects , Statistics, Nonparametric , Tritium/pharmacokinetics
12.
Nat Commun ; 5: 5452, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25406832

ABSTRACT

The evolutionarily conserved peripheral benzodiazepine receptor (PBR), or 18-kDa translocator protein (TSPO), is thought to be essential for cholesterol transport and steroidogenesis, and thus life. TSPO has been proposed as a biomarker of neuroinflammation and a new drug target in neurological diseases ranging from Alzheimer's disease to anxiety. Here we show that global C57BL/6-Tspo(tm1GuWu(GuwiyangWurra))-knockout mice are viable with normal growth, lifespan, cholesterol transport, blood pregnenolone concentration, protoporphyrin IX metabolism, fertility and behaviour. However, while the activation of microglia after neuronal injury appears to be unimpaired, microglia from (GuwiyangWurra)TSPO knockouts produce significantly less ATP, suggesting reduced metabolic activity. Using the isoquinoline PK11195, the ligand originally used for the pharmacological and structural characterization of the PBR/TSPO, and the imidazopyridines CLINDE and PBR111, we demonstrate the utility of (GuwiyangWurra)TSPO knockouts to provide robust data on drug specificity and selectivity, both in vitro and in vivo, as well as the mechanism of action of putative TSPO-targeting drugs.


Subject(s)
Adrenal Glands/diagnostic imaging , Brain/diagnostic imaging , Kidney/diagnostic imaging , Microglia/metabolism , Receptors, GABA/genetics , Adenosine Triphosphate/metabolism , Animals , Behavior, Animal , Cholesterol/metabolism , Fertility/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Positron-Emission Tomography , Pregnenolone/blood , Protoporphyrins/metabolism , Spleen/diagnostic imaging , Testis/diagnostic imaging , Whole Body Imaging
13.
Mol Nutr Food Res ; 58(8): 1750-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24740831

ABSTRACT

Recent research has uncovered the molecular mechanisms responsible for the therapeutic properties of oleanolic acid (OA), its isomer ursolic acid (UA), and derivatives. In particular, recent reports have highlighted the benefits of these compounds in the prevention and treatment of type 2 diabetes and associated life-threatening complications, such as nonalcoholic fatty liver disease, nephropathy, retinopathy, and atherosclerosis. The prevalence of type 2 diabetes is of major concern since it is reaching global epidemic levels. Treatments targeting the signaling pathways altered in type 2 diabetes are being actively investigated, and OA and UA in natural and derivative forms are potential candidates to modulate these pathways. We will explore the findings from in vitro and in vivo studies showing that these compounds: (i) improve insulin signaling and reduce hyperglycemia; (ii) reduce oxidative stress by upregulating anti-oxidants and; (iii) reduce inflammation by inhibiting proinflammatory signaling. We will discuss the molecular mechanisms underpinning these therapeutic properties in this review in order to provide a rationale for the future use of OA, UA, and their derivatives for the prevention and treatment of type 2 diabetes and associated comorbidities.


Subject(s)
Diabetes Complications/prevention & control , Diabetes Mellitus, Type 2/diet therapy , Dietary Supplements , Hypoglycemic Agents/therapeutic use , Models, Biological , Oleanolic Acid/therapeutic use , Triterpenes/therapeutic use , Animals , Diabetes Complications/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/prevention & control , Humans , Hypoglycemic Agents/metabolism , Oleanolic Acid/metabolism , Oxidative Stress , Triterpenes/metabolism , Ursolic Acid
14.
PLoS One ; 9(3): e92618, 2014.
Article in English | MEDLINE | ID: mdl-24675731

ABSTRACT

A low-grade pro-inflammatory state is at the pathogenic core of obesity and type 2 diabetes. We tested the hypothesis that the plant terpenoid compound ginsenoside Rb1 (Rb1), known to exert anti-inflammatory effects, would ameliorate obesity, obesity-associated inflammation and glucose intolerance in the high-fat diet-induced obese mouse model. Furthermore, we examined the effect of Rb1 treatment on central leptin sensitivity and the leptin signaling pathway in the hypothalamus. We found that intraperitoneal injections of Rb1 (14 mg/kg, daily) for 21 days significantly reduced body weight gain, fat mass accumulation, and improved glucose tolerance in obese mice on a HF diet compared to vehicle treatment. Importantly, Rb1 treatment also reduced levels of pro-inflammatory cytokines (TNF-α, IL-6 and/or IL-1ß) and NF-κB pathway molecules (p-IKK and p-IκBα) in adipose tissue and liver. In the hypothalamus, Rb1 treatment decreased the expression of inflammatory markers (IL-6, IL-1ß and p-IKK) and negative regulators of leptin signaling (SOCS3 and PTP1B). Furthermore, Rb1 treatment also restored the anorexic effect of leptin in high-fat fed mice as well as leptin pSTAT3 signaling in the hypothalamus. Ginsenoside Rb1 has potential for use as an anti-obesity therapeutic agent that modulates obesity-induced inflammation and improves central leptin sensitivity in HF diet-induced obesity.


Subject(s)
Ginsenosides/pharmacology , Inflammation/etiology , Inflammation/metabolism , Leptin/metabolism , Obesity/complications , Adiposity/drug effects , Animals , Body Weight/drug effects , Diet, High-Fat , Energy Metabolism/drug effects , Ginsenosides/administration & dosage , Glucose/metabolism , Glucose Tolerance Test , Hormones/blood , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation/drug therapy , Male , Mice , Mice, Obese , Neuropeptides/metabolism , Obesity/etiology , Signal Transduction/drug effects , Time Factors
15.
Brain Res Bull ; 97: 119-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23817050

ABSTRACT

High-fat (HF) diet and obesity are risk factors for a number of mental health problems including depression, cognitive dysfunction, dementia, and neurodegenerative diseases. Histamine H1 receptors (H1Rs) are involved in many of these conditions. This study examined H1R receptor binding density in the brain of male rats fed a high-saturated fat (HF) diet, as well as the effect of docosahexaenoic acid (DHA), galacto-oligosaccharide (GOS) and resistant starch (RS) supplementation of HF diet. Alterations of H1R expression in the post-mortem rat brain were detected by [(3)H]-pyrilamine binding autoradiography. We found that HF diet significantly decreased H1R binding densities in the substantia nigra (SN), caudate putamen (CPu), hypothalamic arcuate nucleus (Arc), ventral tegmental area (VTA), piriform cortex (Pir) and primary motor cortex (M1), compared with low-fat fed rats, and the suppression of receptor binding density ranged from 31% to 48%. Interestingly, supplementing the HF diet with 0.5% n-3 polyunsaturated docosahexaenoic acid (DHA) prevented reduction of H1R binding densities in the SN and CPu. Addition of galacto-oligosaccharide (GOS) and resistant starch (RS) to the diet blunted HF induced reduction of H1R ligand binding in the SN and Pir, respectively. In conclusion this study showed that HF diet can alter H1R binding densities in various brain regions, and many of these changes can be prevented by adding DHA, GOS or RS to the diet.


Subject(s)
Brain/metabolism , Diet, High-Fat , Dietary Fiber/pharmacology , Docosahexaenoic Acids/pharmacology , Receptors, Histamine H1/metabolism , Animals , Brain/diagnostic imaging , Male , Radionuclide Imaging , Rats , Rats, Wistar
16.
Endocrinology ; 154(9): 3130-40, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23751875

ABSTRACT

Chronic inflammation is involved in the pathogenesis of obesity and type 2 diabetes. Recently teasaponin, an extract from tea, has been shown to have antiinflammatory effects. We examined the effect of teasaponin on obesity, inflammation, glucose metabolism, and central leptin sensitivity in obese mice fed a high-fat (HF) diet for 16 weeks. Intraperitoneal injections of teasaponin (10 mg/kg, daily) for 21 days significantly decreased the food intake and body weight of HF diet-induced obese mice. Teasaponin treatment also reduced the protein levels of proinflammatory cytokines (TNF-α, IL-6, and/or IL-1ß) and nuclear factor-κB signaling (phosphorylated inhibitory-κB kinase and phosphorylated inhibitory-κBα) in adipose tissue and the liver. The antiinflammatory effects of teasaponin were associated with improved glycemic status in the treated animals, evidenced by improved glucose tolerance, homeostasis model assessment, and fasting plasma insulin. In the hypothalamus, teasaponin decreased both proinflammatory cytokines and inflammatory signaling in the mediobasal hypothalamus. Teasaponin treatment also enhanced the anorexigenic effect of central leptin administration, restored leptin phosphorylated signal transducer and activator of transcription-3 (p-STAT3) signaling in the arcuate nucleus, and increased hypothalamic expression of the anorexigenic peptide proopiomelanocortin. These results identify a potential novel application for teasaponin as an antiobesity and antiinflammatory agent.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Camellia sinensis/chemistry , Hypothalamus, Middle/drug effects , Leptin/metabolism , Obesity/drug therapy , Plant Extracts/therapeutic use , Tea/chemistry , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Appetite Depressants/administration & dosage , Appetite Depressants/therapeutic use , Diet, High-Fat/adverse effects , Hypothalamus, Middle/immunology , Hypothalamus, Middle/metabolism , Injections, Intraperitoneal , Insulin Resistance , Liver/drug effects , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Plant Extracts/administration & dosage , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/metabolism , Random Allocation , Signal Transduction/drug effects
17.
J Nutr Biochem ; 24(7): 1349-58, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23337348

ABSTRACT

Low levels of docosahexaenoic acid (DHA) have been linked to a number of mental illnesses such as memory loss, depression and schizophrenia. While supplementation of DHA is beneficial in improving memory and cognition, the influence of dietary fats on the neurotransmitters and receptors involved in cognitive function is still not known. The aim of this study was to investigate serotonin receptor (5-HT(1A) and 5-HT2A), cannabinoid receptor (CB1) and gamma-aminobutyric acid type A (GABA(A)) receptor binding densities in the brain of male rats fed a high-saturated-fat (HF) diet, as well as the effect of DHA supplementation on HF diet. Alterations of these receptors in the post-mortem rat brain were detected by [(3)H]-WAY-100635, [(3)H]-ketanserin, [(3)H]-CP-55,940 and [(3)H]-muscimol binding autoradiography, respectively. In the hippocampus, the 5-HT(1A), CB1 and GABA(A) receptor binding densities significantly increased in response to an HF diet, while in the hypothalamus, 5-HT(1A) and CB1 binding densities significantly increased in HF-fed rats. Importantly, DHA supplementation prevented the HF-induced increase of receptors binding density in the hippocampus and hypothalamus. Furthermore, DHA supplementation attenuated 5-HT2A receptor binding density in the caudate putamen, anterior cingulate cortex and medial mammillary nucleus, which was also increased in HF group. This study showed that an HF diet increased 5-HT(1A), 5-HT2A, CB1 and GABA(A) receptor binding densities in the brain regions involved in cognitive function and that dietary DHA can attenuate such alterations. These findings provide insight into the mechanism by which DHA supplementation ameliorates reduced cognitive function associated with an HF diet.


Subject(s)
Brain/drug effects , Dietary Fats/administration & dosage , Docosahexaenoic Acids/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, GABA-A/metabolism , Animals , Brain/metabolism , Male , Protein Binding , Rats , Rats, Wistar
18.
Anal Biochem ; 423(1): 70-7, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22285979

ABSTRACT

The RGD (Arg-Gly-Asp) binding integrins α(v)ß(3) and α(IIb)ß(3) are integral components of various pathological and physiological processes, including tumor angiogenesis, osteoclast function, and thrombus formation. Because of this, there is interest in identifying novel compounds and proteins binding to these receptors as well as investigating the mechanism of these interactions. In this article, we describe the development and validation of competition binding assays for determining the affinity of test compounds to α(v)ß(3) and α(IIb)ß(3) integrin. Assays were successfully developed for each receptor, and the affinity of known compounds was comparable to published results. However, the inability of binding between α(IIb)ß(3) integrin and the labeled echistatin protein ligand to reach equilibrium resulted in an assay that did not meet the assumptions of the competition binding model. Nevertheless, there was good agreement between this assay and known literature values, and intra- and interassay variability was acceptable. Binding by conformation-specific antibodies provided evidence that solid-phase bound α(IIb)ß(3) receptor was in an activated conformation. This study also demonstrated that current models and methods for determining receptor affinity are simplistic and fail to account for common receptor-ligand interactions such as nondissociable interactions and varying receptor activation states.


Subject(s)
Binding, Competitive , Biological Assay/methods , Integrin alphaVbeta3/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Biotinylation , Cell Line, Tumor , Humans , Intercellular Signaling Peptides and Proteins , Isotope Labeling , Kinetics , Oligopeptides/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Binding , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...