Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell Death Dis ; 12(2): 212, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637697

ABSTRACT

Obesity is caused by a long-term imbalance between energy intake and consumption and is regulated by multiple signals. This study investigated the effect of signaling scaffolding protein Gab2 on obesity and its relevant regulation mechanism. Gab2 knockout (KO) and wild-type (WT) mice were fed with a standard diet (SD) or high-fat diet (HFD) for 12 weeks. The results showed that the a high-fat diet-induced Gab2 expression in adipose tissues, but deletion of Gab2 attenuated weight gain and improved glucose tolerance in mice fed with a high-fat diet. White adipose tissue and systemic inflammations were reduced in HFD-fed Gab2 deficiency mice. Gab2 deficiency increased the expression of Ucp1 and other thermogenic genes in brown adipose tissue. Furthermore, the regulation of Gab2 on the mature differentiation and function of adipocytes was investigated in vitro using primary or immortalized brown preadipocytes. The expression of brown fat-selective genes was found to be elevated in differentiated adipocytes without Gab2. The mechanism of Gab2 regulating Ucp1 expression in brown adipocytes involved with its downstream PI3K (p85)-Akt-FoxO1 signaling pathway. Our research suggests that deletion of Gab2 suppresses diet-induced obesity by multiple pathways and Gab2 may be a novel therapeutic target for the treatment of obesity and associated complications.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Energy Metabolism , Obesity/prevention & control , Panniculitis/prevention & control , Adaptor Proteins, Signal Transducing/genetics , Adipose Tissue, Brown/physiopathology , Adipose Tissue, White/physiopathology , Adiposity , Animals , Blood Glucose/metabolism , Cell Line , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Diet, High-Fat , Disease Models, Animal , Forkhead Box Protein O1/metabolism , Insulin Resistance , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Panniculitis/genetics , Panniculitis/metabolism , Panniculitis/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Uncoupling Protein 1/metabolism , Weight Gain
2.
Eur J Pharmacol ; 883: 173371, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32712089

ABSTRACT

Accumulating evidence indicates that adipose tissue inflammation and mitochondrial dysfunction in skeletal muscle are inextricably linked to obesity and insulin resistance. Celastrol, a bioactive compound derived from the root of Tripterygium wilfordii exhibits a number of attributive properties to attenuate metabolic dysfunction in various cellular and animal disease models. However, the underlying therapeutic mechanisms of celastrol in the obesogenic environment in vivo remain elusive. Therefore, the current study investigated the metabolic effects of celastrol on insulin sensitivity, inflammatory response in adipose tissue and mitochondrial functions in skeletal muscle of the high fat diet (HFD)-induced obese rats. Our study revealed that celastrol supplementation at 3 mg/kg/day for 8 weeks significantly reduced the final body weight and enhanced insulin sensitivity of the HFD-fed rats. Celastrol noticeably improved insulin-stimulated glucose uptake activity and increased expression of plasma membrane GLUT4 protein in skeletal muscle. Moreover, celastrol-treated HFD-fed rats showed attenuated inflammatory responses via decreased NF-κB activity and diminished mRNA expression responsible for classically activated macrophage (M1) polarization in adipose tissues. Significant improvement of muscle mitochondrial functions and enhanced antioxidant defense machinery via restoration of mitochondrial complexes I + III linked activity were effectively exhibited by celastrol treatment. Mechanistically, celastrol stimulated mitochondrial biogenesis attributed by upregulation of the adenosine monophosphate-activated protein kinase (AMPK) and sirtuin 1 (SIRT1) signaling pathways. Together, these results further demonstrate heretofore the conceivable therapeutic mechanisms of celastrol in vivo against HFD-induced obesity mediated through attenuation of inflammatory response in adipose tissue and enhanced mitochondrial functions in skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipose Tissue/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Obesity Agents/pharmacology , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Obesity/drug therapy , Panniculitis/prevention & control , Pentacyclic Triterpenes/pharmacology , Sirtuin 1/metabolism , Adipose Tissue/enzymology , Adipose Tissue/physiopathology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diet, High-Fat , Disease Models, Animal , Inflammation Mediators/metabolism , Insulin Resistance , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiopathology , Obesity/enzymology , Obesity/physiopathology , Organelle Biogenesis , Panniculitis/enzymology , Panniculitis/physiopathology , Rats, Sprague-Dawley , Signal Transduction
3.
J Endocrinol Invest ; 43(7): 983-993, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31965518

ABSTRACT

PURPOSE: The alpha7 nicotinic acetylcholine receptor (α7nAChR), involved in the modulation of inflammation and insulin sensitivity, is downregulated in white adipose tissue (WAT) of obese patients. This study aims to test the ability of a selective synthetic α7nAChR agonist, the spirocyclic Δ2-isoxazoline derivative (R)-(-)-ICH3 (ICH3), to counteract acute inflammation and obesity-associated modifications in WAT. METHODS: We employed the LPS-septic shock murine model, human primary adipocytes and diet-induced obese (DIO) mice. Inflammatory factor expression was assessed by ELISA and quantitative real-time PCR. Flow cytometry was employed to define WAT inflammatory infiltrate. Insulin signaling was monitored by quantification of AKT phosphorylation. RESULTS: In the septic shock model, ICH3 revealed antipyretic action and reduced the surge of circulating cytokines. In vitro, ICH3 stimulation (10 µM) preserved viability of human adipocytes, decreased IL-6 mRNA (P < 0.05) and blunted LPS-induced peak of TNFα (P < 0.05) and IL-6 (P < 0.01). Chronic administration of ICH3 to DIO mice was associated with lower numbers of CD8+ T cells (P < 0.05) and to changed WAT expression of inflammatory factors (Hp, P < 0.05; CD301/MGL1, P < 0.01; Arg-1, P < 0.05). As compared to untreated, ICH3 DIO mice exhibited improved insulin signaling in the skeletal muscle (P < 0.01) mirrored by an improved response to glucose load (ipGTT: P < 0.05 at 120 min). CONCLUSIONS: We proved that ICH3 is an anti-inflammatory drug, able to reduce inflammatory cytokines in human adipocytes and to blunt the effects of obesity on WAT inflammatory profile, on glucose tolerance and on tissue insulin sensitivity.


Subject(s)
Adipose Tissue, White/drug effects , Cholinergic Agonists/pharmacology , Fumarates/pharmacology , Obesity/complications , Panniculitis/etiology , Panniculitis/prevention & control , Acetylcholine/agonists , Acetylcholine/analogs & derivatives , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Body Temperature/drug effects , Cells, Cultured , Cholinergic Agonists/therapeutic use , Cytokines/metabolism , Diet, High-Fat , Fumarates/therapeutic use , Glucose/metabolism , Humans , Inflammation/drug therapy , Inflammation/etiology , Inflammation/metabolism , Inflammation Mediators/metabolism , Mice , Mice, Obese , Obesity/drug therapy , Spiro Compounds , alpha7 Nicotinic Acetylcholine Receptor/agonists
4.
J Nutr Biochem ; 72: 108216, 2019 10.
Article in English | MEDLINE | ID: mdl-31476608

ABSTRACT

Impaired intestinal health characterized by a dysbiotic microbial community and a dysfunctional epithelial barrier contributes to host inflammation and metabolic dysfunction in obesity. Fish oil (FO)-derived n-3 polyunsaturated fatty acids have been shown to improve aspects of the obese phenotype; however, their effect on obese intestinal health is unknown. This study aimed to determine the effect of dietary FO on the intestinal microenvironment, including the microbial community and epithelial barrier, in a mouse model of high-fat diet induced obesity and metabolic dysfunction. Male C57BL/6 mice were fed (12 weeks) either a high-fat diet (HF, 60% fat as kcal) or an isocaloric HF supplemented with Menhaden FO (5.3% kcal, HF + FO). 16S rRNA sequencing was used to determine changes in fecal microbiota. Intestinal (ileum and colon) and epididymal adipose tissue RNA was used to assess biomarkers of barrier integrity and inflammatory status, respectively. Serum was used to assess adipokine concentrations and insulin resistance. HF + FO diet altered the fecal microbiota by decreasing the abundance of Firmicutes and increasing the abundance of members of the Bacteroidetes phyla, as well as increasing the abundance of antiobesogenic Akkermansia muciniphila, compared to HF. Intestinal epithelial barrier functions were improved by HF + FO evidenced by increased mRNA expression of tight junction components, antimicrobial defenses and mucus barrier components. HF + FO-fed mice exhibited improvements in homeostatic model assessment of insulin resistance, oral glucose tolerance and serum adipokine concentrations and epididymal mRNA expression (increased adiponectin and decreased leptin) versus HF. HF + FO improved obese intestinal health and attenuated metabolic dysfunction associated with obesity.


Subject(s)
Diet, High-Fat/adverse effects , Fish Oils/pharmacology , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Obesity/diet therapy , Adipokines/blood , Animals , Body Weight/drug effects , Colon/drug effects , Colon/physiology , Dietary Supplements , Eating/drug effects , Fatty Acids, Omega-3/metabolism , Feces/microbiology , Gastrointestinal Microbiome/genetics , Glucose Tolerance Test , Ileum/drug effects , Ileum/physiology , Intestines/physiology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Male , Mice, Inbred C57BL , Obesity/etiology , Panniculitis/etiology , Panniculitis/prevention & control
5.
J Nutr Biochem ; 64: 32-44, 2019 02.
Article in English | MEDLINE | ID: mdl-30428423

ABSTRACT

Saturated fatty acid (SFA) intake is associated with obesity, insulin resistance, and hepatic steatosis, but scant work examines the impact of SFA type upon these outcomes. We tested the hypothesis that an obesogenic diet prepared with medium chain SFA (MCSFA), mostly as lauric acid-derived from coconut oil, reduces obesity-induced outcomes compared to obesogenic diets prepared with increasing amounts long chain SFA (LCSFA), primarily palmitic acid. Mice were fed (16 weeks) a control, low fat diet or obesogenic diets prepared with differing content of MCSFA or LCSFA in which polyunsaturated and monounsaturated fatty acids (PUFA; MUFA) were kept constant. Inclusion of MCSFA in an obesogenic diet prevented hepatic lipid accumulation and lowered indices of insulin resistance. Obesogenic diets reduced hepatic levels of de novo lipogenesis proteins (SCD1 and FASN) but elevated the adipose levels of mRNA for the pro-inflammatory markers Mcp-1 and Tnfα. Lipidomic analysis of plasma indicated that MCSFA intake resulted in a different lipidomic signature than LCSFA intake, prevented elevation of pro-inflammatory ceramides, but elevated concentrations of some lipids associated with elevated cardiovascular disease risk. Intake of the obesogenic diets in an SFA-type dependent manner elevated plasma concentrations of several phosphatidylcholine (PC) lipids having the long chain PUFA (LCPUFA) arachidonic acid (ARA) and docosahexaenoic acid (DHA), altered phospholipid ethers, and changed the triacylglyceryl environments of these LCPUFA. Our data indicate that (1) MCSFA reduce the severity of some obesogenic co-morbidities, (2) SFA-type modulates lipidomic signatures associated with cardiovascular disease and diabetes, and (3) dietary SFA type impacts LCPUFA metabolism.


Subject(s)
Fatty Acids/pharmacology , Lipids/blood , Obesity/metabolism , Animals , Ceramides/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/pharmacology , Hepatitis/etiology , Insulin Resistance , Lipogenesis/drug effects , Male , Mice, Inbred C57BL , Obesity/etiology , Panniculitis/etiology , Panniculitis/prevention & control
6.
Gut ; 68(3): 453-464, 2019 03.
Article in English | MEDLINE | ID: mdl-30064988

ABSTRACT

OBJECTIVE: The consumption of fruits is strongly associated with better health and higher bacterial diversity in the gut microbiota (GM). Camu camu (Myrciaria dubia) is an Amazonian fruit with a unique phytochemical profile, strong antioxidant potential and purported anti-inflammatory potential. DESIGN: By using metabolic tests coupled with 16S rRNA gene-based taxonomic profiling and faecal microbial transplantation (FMT), we have assessed the effect of a crude extract of camu camu (CC) on obesity and associated immunometabolic disorders in high fat/high sucrose (HFHS)-fed mice. RESULTS: Treatment of HFHS-fed mice with CC prevented weight gain, lowered fat accumulation and blunted metabolic inflammation and endotoxaemia. CC-treated mice displayed improved glucose tolerance and insulin sensitivity and were also fully protected against hepatic steatosis. These effects were linked to increased energy expenditure and upregulation of uncoupling protein 1 mRNA expression in the brown adipose tissue (BAT) of CC-treated mice, which strongly correlated with the mRNA expression of the membrane bile acid (BA) receptor TGR5. Moreover, CC-treated mice showed altered plasma BA pool size and composition and drastic changes in the GM (eg, bloom of Akkermansia muciniphila and a strong reduction of Lactobacillus). Germ-free (GF) mice reconstituted with the GM of CC-treated mice gained less weight and displayed higher energy expenditure than GF-mice colonised with the FM of HFHS controls. CONCLUSION: Our results show that CC prevents visceral and liver fat deposition through BAT activation and increased energy expenditure, a mechanism that is dependent on the GM and linked to major changes in the BA pool size and composition.


Subject(s)
Energy Metabolism/physiology , Fruit/chemistry , Gastrointestinal Microbiome/drug effects , Obesity/prevention & control , Animals , Ascorbic Acid/therapeutic use , Blood Glucose/metabolism , Endotoxemia/prevention & control , Fatty Liver/microbiology , Fatty Liver/physiopathology , Fatty Liver/prevention & control , Fecal Microbiota Transplantation , Homeostasis/physiology , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/microbiology , Obesity/physiopathology , Panniculitis/prevention & control , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
7.
J Physiol Biochem ; 74(4): 647-654, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30232707

ABSTRACT

To fight against metabolic disorders such as insulin resistance, new alimentary behaviors are developed. For instance, hyperproteined, gluten-free, or collagen-enriched diets could be preconized in order to reduce the consequences of obesity. In this aim, this study evaluates the potential effects of warm sea fish collagen peptides (Naticol®) on representative metabolic and inflammatory parameters. For that, male C57Bl6/J mice fed with either a chow- (CD) or high-fat diet (HFD) were submitted or not to specific collagen peptides in drinking water (4 g/kg bw/d) for 20 weeks. Weight, body composition, glucose tolerance, and insulin sensitivity were followed up. Effects of fish collagen peptides on various blood parameters reflecting the metabolism status were also measured (free fatty acids, triglycerides, cholesterol, hormones) together with adipocyte inflammation. Results showed that HFD-fed mice supplemented by fish collagen peptides exhibited a significant lower increase in body weight as soon as the twelfth week of treatment whereas no effect of the peptide was observed in CD fed mice. In line with this result, a weaker increase in fat mass in HFD-fed mice supplemented with Naticol® at both 9 and 18 weeks of treatment was also observed. In spite of this resistance to obesity promoted by fish collagen peptides treatment, no difference in glucose tolerance was found between groups whereas mice treated with Naticol® exhibited a lower basal glycemia. Also, even if no effect of the treatment on adipocyte lipolysis was found, a decrease of inflammatory cytokines was retrieved in collagen-supplemented group arguing for a potential better insulin sensitivity. Altogether, these results need to be completed but are the first describing a benefic role of warm sea fish collagen peptides in a context of metabolic disease paving the route for a potential utilization in human obesity-associated disorders.


Subject(s)
Anti-Obesity Agents/therapeutic use , Collagen/therapeutic use , Dietary Supplements , Fish Proteins, Dietary/therapeutic use , Insulin Resistance , Obesity/therapy , Peptide Fragments/therapeutic use , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/metabolism , Apelin/agonists , Apelin/genetics , Apelin/metabolism , Collagen/adverse effects , Collagen/chemistry , Collagen/metabolism , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements/adverse effects , Fish Proteins, Dietary/adverse effects , Fish Proteins, Dietary/chemistry , Fish Proteins, Dietary/metabolism , Gene Expression Regulation , Glucose Intolerance/etiology , Glucose Intolerance/immunology , Glucose Intolerance/prevention & control , Lipolysis , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology , Panniculitis/etiology , Panniculitis/immunology , Panniculitis/prevention & control , Peptide Fragments/adverse effects , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Weight Gain
8.
J Nutr Biochem ; 58: 1-16, 2018 08.
Article in English | MEDLINE | ID: mdl-29621669

ABSTRACT

Strategies to reduce obesity have become public health priorities as the prevalence of obesity has risen in the United States and around the world. While the anti-inflammatory and hypotriglyceridemic properties of long-chain omega-3 polyunsaturated fatty acids (n-3 PUFAs) are well known, their antiobesity effects and efficacy against metabolic syndrome, especially in humans, are still under debate. In animal models, evidence consistently suggests a role for n-3 PUFAs in reducing fat mass, particularly in the retroperitoneal and epididymal regions. In humans, however, published research suggests that though n-3 PUFAs may not aid weight loss, they may attenuate further weight gain and could be useful in the diet or as a supplement to help maintain weight loss. Proposed mechanisms by which n-3 PUFAs may work to improve body composition and counteract obesity-related metabolic changes include modulating lipid metabolism; regulating adipokines, such as adiponectin and leptin; alleviating adipose tissue inflammation; promoting adipogenesis and altering epigenetic mechanisms.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Metabolic Syndrome/diet therapy , Obesity/diet therapy , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Anti-Obesity Agents/pharmacology , Body Composition/drug effects , Exercise , Humans , Insulin Resistance , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Panniculitis/diet therapy , Panniculitis/metabolism , Panniculitis/prevention & control , Vegetables/chemistry
9.
J Nutr Biochem ; 55: 12-25, 2018 05.
Article in English | MEDLINE | ID: mdl-29331880

ABSTRACT

Indole-3-carbinol (I3C), found in Brassica family vegetables, exhibits antioxidant, anti-inflammatory, and anti-cancerous properties. Here, we aimed to evaluate the preventive effects of I3C against ethanol (EtOH)-induced liver injury and study the protective mechanism(s) by using the well-established chronic-plus-binge alcohol exposure model. The preventive effects of I3C were evaluated by conducting various histological, biochemical, and real-time PCR analyses in mouse liver, adipose tissue, and colon, since functional alterations of adipose tissue and intestine can also participate in promoting EtOH-induced liver damage. Daily treatment with I3C alleviated EtOH-induced liver injury and hepatocyte apoptosis, but not steatosis, by attenuating elevated oxidative stress, as evidenced by the decreased levels of hepatic lipid peroxidation, hydrogen peroxide, CYP2E1, NADPH-oxidase, and protein acetylation with maintenance of mitochondrial complex I, II, and III protein levels and activities. I3C also restored the hepatic antioxidant capacity by preventing EtOH-induced suppression of glutathione contents and mitochondrial aldehyde dehydrogenase-2 activity. I3C preventive effects were also achieved by attenuating the increased levels of hepatic proinflammatory cytokines, including IL1ß, and neutrophil infiltration. I3C also attenuated EtOH-induced gut leakiness with decreased serum endotoxin levels through preventing EtOH-induced oxidative stress, apoptosis of enterocytes, and alteration of tight junction protein claudin-1. Furthermore, I3C alleviated adipose tissue inflammation and decreased free fatty acid release. Collectively, I3C prevented EtOH-induced liver injury via attenuating the damaging effect of ethanol on the gut-liver-adipose tissue axis. Therefore, I3C may also have a high potential for translational research in treating or preventing other types of hepatic injury associated with oxidative stress and inflammation.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic/pathology , Chemical and Drug Induced Liver Injury, Chronic/prevention & control , Ethanol/adverse effects , Indoles/pharmacology , Alcoholism/pathology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Colon/drug effects , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/pathology , Male , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/pathology , Oxidative Stress/drug effects , Panniculitis/pathology , Panniculitis/prevention & control
10.
Metab Syndr Relat Disord ; 15(9): 450-457, 2017 11.
Article in English | MEDLINE | ID: mdl-28934021

ABSTRACT

BACKGROUND: Perivascular adipose tissue (PVAT) can regulate vascular homeostasis by secreting various adipokines. This study investigated the effects of PVAT browning on its endocrine function. METHODS: In the first section of our study, male Sprague-Dawley rats were randomly divided into cold exposure (8°C) and 24°C acclimation groups. After cold exposure for 7 days, interscapular brown adipose tissue (iBAT), subcutaneous white adipose tissue, thoracic aortic PVAT, and abdominal aortic PVAT (aPVAT) were harvested for histological and brown marker gene expression analysis. In the second part, male rats were fed a high fat diet (HFD) for 10 weeks. In the 11th week, the rats were treated with or without cold exposure. After 14-day cold exposure, aPVAT was collected for histological, gene, and protein expression analysis. RESULTS: Cold exposure had a browning effect on aPVAT by increasing UCP-1 and PGC-1α expression levels. After HFD feeding for 10 weeks, 14-day cold exposure was still able to induce aPVAT browning. Compared with thermoneutrality acclimation rats, TNF-α, IL-6, and p-p65 expression levels were significantly lower in aPVAT from HFD-fed rats with cold exposure. In contrast, p-AMPK expression levels were increased in aPVAT from HFD-fed rats with cold exposure. CONCLUSIONS: Our study demonstrated that browning of aPVAT in HFD-fed rats lowered the pro-inflammatory adipokine expression levels and activated AMPK.


Subject(s)
Abdominal Fat/physiology , Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Cell Transdifferentiation/physiology , Cold Temperature , Inflammation/prevention & control , Panniculitis/prevention & control , Adipokines/metabolism , Animals , Aorta, Abdominal , Male , Rats , Rats, Sprague-Dawley
11.
J Biol Chem ; 292(34): 13959-13969, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28698385

ABSTRACT

Adipose tissue inflammation has been linked to metabolic diseases such as obesity and type 2 diabetes. However, the molecules that mediate inflammation in adipose tissue have not been addressed. Although retinoic acid receptor-related orphan receptor α (RORα) is known to be involved in the regulation of inflammatory response in some tissues, its role is largely unknown in adipose tissue. Conversely, it is known that endoplasmic reticulum (ER) stress and unfolding protein response (UPR) signaling affect the inflammatory response in obese adipose tissue, but whether RORα regulates these processes remains unknown. In this study, we investigate the link between RORα and adipose tissue inflammation. We showed that the inflammatory response in macrophages or 3T3-L1 adipocytes stimulated by lipopolysaccharide, as well as adipose tissue in obese mice, markedly increased the expression of RORα. Adenovirus-mediated overexpression of RORα or treatment with the RORα-specific agonist SR1078 enhanced the expression of inflammatory cytokines and increased the number of infiltrated macrophages into adipose tissue. Furthermore, SR1078 up-regulated the mRNA expression of ER stress response genes and enhanced phosphorylations of two of the three mediators of major UPR signaling pathways, PERK and IRE1α. Finally, we found that alleviation of ER stress using a chemical chaperone followed by the suppression of RORα induced inflammation in adipose tissue. Our data suggest that RORα-induced ER stress response potentially contributes to the adipose tissue inflammation that can be mitigated by treatment with chemical chaperones. The relationships established here between RORα expression, inflammation, and UPR signaling may have implications for therapeutic targeting of obesity-related metabolic diseases.


Subject(s)
Adipocytes, White/metabolism , Endoplasmic Reticulum Stress , Macrophages/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Panniculitis/metabolism , Signal Transduction/drug effects , Unfolded Protein Response , 3T3-L1 Cells , Adipocytes, White/drug effects , Adipocytes, White/immunology , Adipocytes, White/pathology , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Insulin Resistance , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Obesity/drug therapy , Obesity/physiopathology , Panniculitis/immunology , Panniculitis/pathology , Panniculitis/prevention & control , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , RAW 264.7 Cells , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Unfolded Protein Response/drug effects
12.
J Nutr Biochem ; 43: 27-35, 2017 05.
Article in English | MEDLINE | ID: mdl-28193580

ABSTRACT

The objective of this study was to determine if astaxanthin (ASTX), a xanthophyll carotenoid, can prevent obesity-associated metabolic abnormalities, inflammation and fibrosis in diet-induced obesity (DIO) and nonalcoholic steatohepatitis (NASH) mouse models. Male C57BL/6J mice were fed a low-fat (6% fat, w/w), a high-fat/high-sucrose control (HF/HS; 35% fat, 35% sucrose, w/w), or a HF/HS containing ASTX (AHF/HS; 0.03% ASTX, w/w) for 30 weeks. To induce NASH, another set of mice was fed a HF/HS diet containing 2% cholesterol (HF/HS/HC) a HF/HS/HC with 0.015% ASTX (AHF/HS/HC) for 18 weeks. Compared to LF, HF/HS significantly increased plasma total cholesterol, triglyceride and glucose, which were lowered by ASTX. ASTX decreased hepatic mRNA levels of markers of macrophages and fibrosis in both models. The effect of ASTX was more prominent in NASH than DIO mice. In epididymal fat, ASTX also decreased macrophage infiltration and M1 macrophage marker expression, and inhibited hypoxia-inducible factor 1-α and its downstream fibrogenic genes in both mouse models. ASTX significantly decreased tumor necrosis factor α mRNA in the splenocytes from DIO mice upon lipopolysaccharides stimulation compared with those from control mice fed an HF/HS diet. Additionally, ASTX significantly elevated the levels of genes that regulate fatty acid ß-oxidation and mitochondrial biogenesis in the skeletal muscle compared with control obese mice, whereas no differences were noted in adipose lipogenic genes. Our results indicate that ASTX inhibits inflammation and fibrosis in the liver and adipose tissue and enhances the skeletal muscle's capacity for mitochondrial fatty acid oxidation in obese mice.


Subject(s)
Adipose Tissue/pathology , Liver/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/complications , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Dietary Supplements , Disease Models, Animal , Fibrosis/prevention & control , Gene Expression Regulation/drug effects , Lipids/blood , Lipids/genetics , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , Obesity/etiology , Obesity/prevention & control , Panniculitis/metabolism , Panniculitis/pathology , Panniculitis/prevention & control , Xanthophylls/pharmacology
13.
Biochem Biophys Res Commun ; 484(3): 480-485, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28089867

ABSTRACT

Obesity is associated with chronic inflammation of adipose tissue and causes development of type 2 diabetes. M1 macrophage population was increased in adipose tissue of obese mouse. M1 macrophages induce insulin resistance through the secretion of proinflammatory cytokines. Our previous studies demonstrated that salmon cartilage proteoglycan (PG) suppresses excess inflammation in various mouse inflammatory diseases. In this study, we examined the effect of PG on type 2 diabetes using high-fat-diet (HFD) induced obese mouse model. Oral PG administration enhanced the population of small adipocytes (area less than 1000 µm2) without body and tissue weight gain. In addition, PG administration suppressed mRNA expression of TNF-α, IL-6 and CXCL2 in adipose tissue. The proportion of M1 macrophages was decreased by PG administration. In addition, PG administration suppressed hyperglycemia after intraperitoneal glucose injection. Fasted serum insulin level was decreased in PG-administered mice. Moreover, insulin-stimulated phosphorylation of Akt was enhanced in the liver and gastrocnemius skeletal muscle of PG-administered mice. These data suggested that PG administration improves hyperglycemia and insulin sensitivity in obese mice by modulation of M1 macrophages which secrete proinflammatory cytokines in adipose tissue and activation of Akt in liver and skeletal muscle.


Subject(s)
Cartilage/chemistry , Inflammation Mediators/immunology , Obesity/immunology , Panniculitis/immunology , Panniculitis/prevention & control , Proteoglycans/administration & dosage , Administration, Oral , Animals , Biological Products/administration & dosage , Cytokines/immunology , Diet, High-Fat , Dose-Response Relationship, Drug , Inflammation , Insulin Resistance/immunology , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Salmon , Treatment Outcome
14.
Arterioscler Thromb Vasc Biol ; 37(3): 466-475, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28062496

ABSTRACT

OBJECTIVE: Obesity is associated with insulin resistance and adipose tissue inflammation. Reactive oxygen species (ROS) increase in adipose tissue during the development of obesity. We previously showed that in response to excess nutrients like glucose and palmitate, adipocytes generated ROS via NADPH oxidase (NOX) 4, the major adipocyte isoform, instead of using mitochondrial oxidation. However, the role of NOX4-derived ROS in the development of whole body insulin resistance, adipocyte inflammation, and recruitment of macrophages to adipose tissue during the development of obesity is unknown. APPROACH AND RESULTS: In this study, control C57BL/6 mice and mice in which NOX4 has been deleted specifically in adipocytes were fed a high-fat, high-sucrose diet. During the development of obesity in control mice, adipocyte NOX4 and pentose phosphate pathway activity were transiently increased. Primary adipocytes differentiated from mice with adipocytes deficient in NOX4 showed resistance against high glucose or palmitate-induced adipocyte inflammation. Mice with adipocytes deficient in NOX4 showed a delayed onset of insulin resistance during the development of obesity, with an initial reduction in adipose tissue inflammation that normalized with prolonged high-fat, high-sucrose feeding. CONCLUSIONS: These findings imply that NOX4-derived ROS may play a role in the onset of insulin resistance and adipose tissue inflammation. As such, therapeutics targeting NOX4-mediated ROS production could be effective in preventing obesity-associated conditions, such as insulin resistance.


Subject(s)
Adipocytes/enzymology , Adipose Tissue/enzymology , Insulin Resistance , NADPH Oxidases/deficiency , Obesity/enzymology , Panniculitis/prevention & control , Animals , Cells, Cultured , Diet, High-Fat , Dietary Sucrose , Disease Models, Animal , Genotype , Hepatitis/enzymology , Hepatitis/genetics , Hepatitis/prevention & control , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4 , NADPH Oxidases/genetics , Obesity/genetics , Panniculitis/enzymology , Panniculitis/genetics , Pentose Phosphate Pathway , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction
15.
Nutr Metab Cardiovasc Dis ; 27(3): 249-259, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28062181

ABSTRACT

BACKGROUND AND AIMS: Epicatechin (EC) intake has been suggested to be beneficial for the prevention of cardiovascular disorders, and it is well known that adipose tissue inflammation is one of the major risk factors for coronary heart diseases. The purpose of the present study was to determine the in vitro and in vivo effects of EC on adipose tissue inflammation and obesity. METHODS AND RESULTS: DNA microarray analysis was performed to evaluate the effects of EC on gene expression in adipocytes co-cultured with bacterial endotoxin-stimulated macrophages. To determine the in vivo effects of the catechin, C57BL/6 mice were fed either a high-fat diet (HFD) or HFD combined with EC, and metabolic changes were observed EC suppressed the expression of many inflammatory genes in the adipocytes co-cultured with endotoxin-stimulated macrophages. Specifically, EC markedly suppressed chemokine (CC motif) ligand 19 (CCL19) expression. The target cell of EC appeared to macrophages. The in vivo study indicated that mice fed the EC-supplemented HFD were protected from diet-induced obesity and insulin resistance. Accordingly, the expression levels of genes associated with inflammation in adipose tissue and in the liver were downregulated in this group of mice. CONCLUSIONS: EC exerts beneficial effects for the prevention of adipose tissue inflammation and insulin resistance. Since we previously reported that mice deficient in the CCL19 receptor were protected from diet-induced obesity and insulin resistance, it can be concluded that the beneficial effects of EC could be mediated, at least in part, by marked suppression of CCL19 expression.


Subject(s)
Adipocytes/drug effects , Adipose Tissue/drug effects , Anti-Inflammatory Agents/pharmacology , Catechin/pharmacology , Chemokine CCL19/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance , Obesity/prevention & control , Panniculitis/prevention & control , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Chemokine CCL19/genetics , Coculture Techniques , Disease Models, Animal , Down-Regulation , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Panniculitis/etiology , Panniculitis/genetics , Panniculitis/metabolism , RAW 264.7 Cells , Time Factors
16.
Aging (Albany NY) ; 8(1): 178-91, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26837433

ABSTRACT

Aging is commonly associated with low-grade adipose inflammation, which is closely linked to insulin resistance. Ghrelin is the only circulating orexigenic hormone which is known to increase obesity and insulin resistance. We previously reported that the expression of the ghrelin receptor, growth hormone secretagogue receptor (GHS-R), increases in adipose tissues during aging, and old Ghsr(-/-) mice exhibit a lean and insulin-sensitive phenotype. Macrophages are major mediators of adipose tissue inflammation, which consist of pro-inflammatory M1 and anti-inflammatory M2 subtypes. Here, we show that in aged mice, GHS-R ablation promotes macrophage phenotypical shift toward anti-inflammatory M2. Old Ghsrp(-/-) mice have reduced macrophage infiltration, M1/M2 ratio, and pro-inflammatory cytokine expression in white and brown adipose tissues. We also found that peritoneal macrophages of old Ghsrp(-/-) mice produce higher norepinephrine, which is in line with increased alternatively-activated M2 macrophages. Our data further reveal that GHS-R has cell-autonomous effects in macrophages, and GHS-R antagonist suppresses lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. Collectively, our studies demonstrate that ghrelin signaling has an important role in macrophage polarization and adipose tissue inflammation during aging. GHS-R antagonists may serve as a novel and effective therapeutic option for age-associated adipose tissue inflammation and insulin resistance.


Subject(s)
Aging/metabolism , Intra-Abdominal Fat/metabolism , Macrophages, Peritoneal/metabolism , Panniculitis/metabolism , Receptors, Ghrelin/metabolism , Age Factors , Aging/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Cell Plasticity , Genetic Predisposition to Disease , Hormone Antagonists/pharmacology , Inflammation Mediators/metabolism , Insulin Resistance , Intra-Abdominal Fat/drug effects , Lipolysis , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Panniculitis/genetics , Panniculitis/prevention & control , Phenotype , RAW 264.7 Cells , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/genetics
17.
Diabetes ; 65(4): 956-66, 2016 04.
Article in English | MEDLINE | ID: mdl-26718500

ABSTRACT

Obesity is associated with an increased risk for the development of type 2 diabetes and vascular complications. Advanced glycation end products are increased in adipose tissue and have been associated with insulin resistance, vascular dysfunction, and inflammation of adipose tissue. Here, we report that delayed intervention with pyridoxamine (PM), a vitamin B6 analog that has been identified as an antiglycating agent, protected against high-fat diet (HFD)-induced body weight gain, hyperglycemia, and hypercholesterolemia, compared with mice that were not treated. In both HFD-induced and db/db obese mice, impaired glucose metabolism and insulin resistance were prevented by PM supplementation. PM inhibited the expansion of adipose tissue and adipocyte hypertrophy in mice. In addition, adipogenesis of murine 3T3-L1 and human Simpson-Golabi-Behmel Syndrome preadipocytes was dose- and time-dependently reduced by PM, as demonstrated by Oil Red O staining and reduced expression of adipogenic differentiation genes. No ectopic fat deposition was found in the liver of HFD mice. The high expression of proinflammatory genes in visceral adipose tissue of the HFD group was significantly attenuated by PM. Treatment with PM partially prevented HFD-induced mild vascular dysfunction. Altogether, these findings highlight the potential of PM to serve as an intervention strategy in obesity.


Subject(s)
Inflammation/prevention & control , Insulin Resistance , Obesity/drug therapy , Panniculitis/prevention & control , Pyridoxamine/administration & dosage , 3T3-L1 Cells , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Cells, Cultured , Diet, High-Fat , Drug Administration Schedule , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Gigantism/metabolism , Gigantism/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Humans , Inflammation/metabolism , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/metabolism , Panniculitis/metabolism , Time-to-Treatment
18.
J Endocrinol ; 227(1): 25-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26264980

ABSTRACT

A decrease in serum estrogen levels in menopause is closely associated with the development of visceral obesity and the onset of type 2 diabetes in women. In the present study, we demonstrated the therapeutic effects of the novel DPP4 inhibitor, teneligliptin, on the features of postmenopausal obesity in mice. In the control group, female C57BL/6 mice were sham-operated and maintained on a standard diet. In the postmenopausal obese group, ovariectomized (OVX) mice were maintained on a high-fat diet, and were referred to as OVX-HF. In the treated group, teneligliptin at 60 mg/kg per day was administrated to OVX-HF, and were referred to as Tene. After a 12-week food challenge, the metabolic phenotypes of these mice were analyzed. Body weight, fat accumulation, and glucose intolerance were greater in OVX-HF than in control, while these abnormalities were markedly improved without alterations in calorie intake in Tene. Teneligliptin effectively ameliorated the characteristics of metabolic abnormalities associated with postmenopausal obesity. Regarding chronic inflammation in visceral adipose tissue, the numbers of F4/80(+)CD11c(+)CD206(-) M1-macrophages in flow cytometry, crown-like structure formation in immunohistochemistry, and proinflammatory cytokine expression were significantly attenuated in Tene. Hepatic steatosis was also markedly improved. Furthermore, decreased energy consumption in the dark and light phases, reduced locomotor activity in the dark phase, and lowered core body temperature in OVX-HF were ameliorated in Tene. Since obesity and reduced energy metabolism are a common physiology of menopause, teneligliptin appears to be beneficial as a treatment for type 2 diabetes in postmenopausal obesity.


Subject(s)
Aging , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Disease Models, Animal , Obesity, Abdominal/complications , Panniculitis/prevention & control , Pyrazoles/therapeutic use , Thiazolidines/therapeutic use , Animals , Behavior, Animal/drug effects , Body Temperature Regulation/drug effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Female , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Liver/drug effects , Liver/immunology , Liver/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Motor Activity/drug effects , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity, Abdominal/etiology , Obesity, Abdominal/physiopathology , Ovariectomy , Panniculitis/etiology
19.
J Nutr Biochem ; 26(9): 960-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25997866

ABSTRACT

Fish oil improves obesity and its comorbidities, but its mechanisms of action remain unknown. We evaluate the effects of a diet rich in fish oil in white adipose tissue (WAT) inflammation pathways, renin-angiotensin system (RAS) and mitogen-activated protein kinases (MAPKs). To achieve our aims, four groups of male C57BL/6 mice were fed different diets: standard chow diet (SC; 10% energy from fat), SC+fish oil diet (SC-FO; 10% energy from fat), high-fat lard diet (HF-L; 50% energy from lard) and HF fish oil diet (HF-FO; 50% energy from fish oil). We evaluated body mass, epididymal fat pad mass, food intake and glucose tolerance. In WAT, we assessed adipocyte hypertrophy, monocyte chemotactic protein-1 immunofluorescence, and gene and protein expression of insulin signaling, inflammation, MAPKs, RAS, peroxisome proliferator-activated receptors (PPARs) and AMP-activated protein kinase (AMPK). In relation to the results, the HF-L group, as expected, showed elevated body mass and adiposity, glucose intolerance and hypertrophied adipocytes. In WAT, we found a defect in insulin signaling, infiltration of macrophages and inflammatory markers with the associated activation of MAPKs and local RAS. On the contrary, the HF-FO group did not present increased body mass, adiposity or glucose intolerance. In this group, insulin signaling, macrophage infiltration and inflammation were reduced in WAT in comparison with the HF-L group. We also observed decreases of MAPKs and local RAS and elevation of PPAR and AMPK. In summary, fish oil activates PPAR (the three isoforms) and AMPK, decreases WAT insulin resistance and inflammation, and inhibits MAPK and RAS pathways activation.


Subject(s)
Adipose Tissue, White/immunology , Adiposity , Dietary Supplements , Fish Oils/therapeutic use , Insulin Resistance , Obesity/prevention & control , Panniculitis/prevention & control , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Diet, High-Fat/adverse effects , Fish Oils/administration & dosage , Glucose Intolerance/etiology , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Glucose Intolerance/prevention & control , Hypertrophy , Inflammation Mediators/metabolism , MAP Kinase Signaling System , Macrophage Activation , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Panniculitis/etiology , Panniculitis/immunology , Panniculitis/metabolism , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Random Allocation , Weight Gain
20.
Am J Physiol Endocrinol Metab ; 308(8): E621-30, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25714672

ABSTRACT

We hypothesized that carbon monoxide (CO) might suppress chronic inflammation, which led to metabolic disturbances. Ovariectomy (OVX) was performed in mice to mimic chronic inflammation secondary to loss of ovarian function. OVX increased fat mass and the infiltration of highly inflammatory CD11c cells into adipose tissue (AT), resulting in a disturbance of glucose metabolism. Treatment of CO attenuated these; CO decreased recruitment of CD11c-expressing cells in AT and reduced expression of CD11c in bone marrow-derived macrophages, protecting them from M1 polarization. Upregulated cGMP and decreased reactive oxygen species were responsible for the inhibitory activity of CO on CD11c expression; knockdown of soluble guanylate cyclase or heme oxygenase-1 using small interfering RNAs reduced this inhibition substantially. Improved OVX-induced insulin resistance (IR) by CO was highly associated with its activity to attenuate AT inflammation. Our results suggest a therapeutic value of CO to treat postmenopausal IR by reducing AT inflammation.


Subject(s)
Adipose Tissue, White/drug effects , Aging , Antimetabolites/pharmacology , Carbon Monoxide/pharmacology , Insulin Resistance , Macrophages/drug effects , Panniculitis/prevention & control , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity/drug effects , Animals , Cells, Cultured , Cyclic GMP/agonists , Cyclic GMP/metabolism , Female , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Injections, Intraperitoneal , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Organometallic Compounds/administration & dosage , Organometallic Compounds/pharmacology , Organometallic Compounds/therapeutic use , Ovariectomy/adverse effects , Panniculitis/immunology , Panniculitis/metabolism , Panniculitis/pathology , Prodrugs/administration & dosage , Prodrugs/pharmacology , Prodrugs/therapeutic use , RNA Interference , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL
...