Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Science ; 377(6601): 47-56, 2022 07.
Article in English | MEDLINE | ID: mdl-35771919

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) kinase controls growth in response to nutrients, including the amino acid leucine. In cultured cells, mTORC1 senses leucine through the leucine-binding Sestrin proteins, but the physiological functions and distribution of Sestrin-mediated leucine sensing in mammals are unknown. We find that mice lacking Sestrin1 and Sestrin2 cannot inhibit mTORC1 upon dietary leucine deprivation and suffer a rapid loss of white adipose tissue (WAT) and muscle. The WAT loss is driven by aberrant mTORC1 activity and fibroblast growth factor 21 (FGF21) production in the liver. Sestrin expression in the liver lobule is zonated, accounting for zone-specific regulation of mTORC1 activity and FGF21 induction by leucine. These results establish the mammalian Sestrins as physiological leucine sensors and reveal a spatial organization to nutrient sensing by the mTORC1 pathway.


Subject(s)
Diet , Leucine , Liver , Mechanistic Target of Rapamycin Complex 1 , Sestrins , Adipose Tissue, White/enzymology , Animals , Leucine/metabolism , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Sestrins/metabolism , Signal Transduction
2.
J Endocrinol ; 248(1): 87-93, 2021 01.
Article in English | MEDLINE | ID: mdl-33112799

ABSTRACT

Obesity is classically associated with low serum total and free 25(OH)D. Hypotheses have been advanced to explain this observation but mechanisms remain poorly understood, and notably priming events that could explain such association. We investigated the impact of short-term high fat (HF) diet to investigate early events occurring in vitamin D metabolism. Male C57BL/6J mice were fed with a control diet (control group) and HF diet for 4 days. HF fed mice displayed similar body weight to control mice but significantly increased adiposity, together with a decrease of free 25(OH)D concentrations, which could be explained at least in part by a decrease of Cyp2r1 and Cyp3a11 expression in the liver. An increase of 1,25(OH)2D concentration was also observed and could be explained by a decrease of Cyp24a1 expression observed in the kidney. In white adipose tissue (WAT), no modification of vitamin D metabolites quantity detected by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Nevertheless, an increase of Cyp2r1 and Cyp27a1 mRNA expression and a decrease of Cyp27b1 mRNA expression could suggest a possible storage of 25(OH)D in WAT at long-term. Our data are supportive of an active role of HF diet in mediating a priming effect leading the well-established perturbation of the vitamin D metabolism associated with obesity, including a decrease of free 25(OH)D and modulation of expression of genes involved in vitamin D metabolism.


Subject(s)
Diet, High-Fat/adverse effects , Obesity/enzymology , Vitamin D/analogs & derivatives , Adipose Tissue, White/enzymology , Animals , Cholecalciferol/blood , Gene Expression Profiling , Kidney/enzymology , Liver/enzymology , Male , Mice, Inbred C57BL , Obesity/etiology , Vitamin D/metabolism
3.
Acta Pharmacol Sin ; 42(6): 964-974, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32934347

ABSTRACT

Beige adipocytes have been considered as a potential strategy in anti-obesity therapy because of its thermogenic capacity. AMP-activated protein kinase (AMPK) plays important roles in regulating adipose tissue function. C29 is a novel pyrazolone derivative with AMPK activity. In the current study, we investigated the role of C29 in the regulation of thermogenesis using differentiated adipocytes and diet-induced obese mice, and explored the mechanisms that might be involved in energy expenditure via adipocyte AMPK activation. We showed that treatment with C29 (2.5-10 µM) concentration-dependently increased thermogenesis in differentiated preadipocytes separated from inguinal white adipose tissue (iWAT), evidenced by increased expression levels of thermogenesis markers such as Ucp1, Pgc-1α, Dio2, Prdm16, Cox7a1, Cox8b, Elovl3, and Cidea, fatty acid oxidation (FAO) genes including Cpt1a, Lcad and Pparα, as well as beige-selective genes such as Cd137, Tmem26, Slc27a1, and Tbx1. In high-fat diet (HFD)-fed mice, oral administration of C29 (30 mg·kg-1·day-1) for 9 weeks alleviated HFD-induced obesity, promoted energy expenditure and modulated iWAT browning. However, these effects were not observed in adipose-specific AMPKα1/α2 knockout (AKO) mice following C29 administration. Together, this study demonstrates that C29 regulates energy balance via adipocyte AMPK. Our findings show that the discovery of AMPK activators that specifically target adipose tissue may have therapeutic potential for treating obesity-related metabolic diseases.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipose Tissue, White/drug effects , Enzyme Activators/therapeutic use , Obesity/drug therapy , Pyrazolones/therapeutic use , Adipocytes/drug effects , Adipose Tissue, Beige/enzymology , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/enzymology , Adipose Tissue, White/metabolism , Animals , Body Temperature/drug effects , Cell Differentiation/drug effects , Diet, High-Fat , Insulin Resistance/physiology , Male , Mice, Inbred C57BL , Obesity/enzymology , Obesity/metabolism , Thermogenesis/drug effects
4.
Am J Physiol Heart Circ Physiol ; 320(1): H432-H446, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33185110

ABSTRACT

Following cardiac injury, increased adrenergic drive plays an important role in compensating for reduced cardiac function. However, chronic excess adrenergic stimulation can be detrimental to cardiac pathophysiology and can also affect other organs including adipose tissue, leading to increased lipolysis. Interestingly, inhibition of adipose triglyceride lipase (ATGL), a rate-limiting enzyme in lipolysis, in adipocytes ameliorates cardiac dysfunction in a heart failure model. Thus, we investigated whether inhibition of adipocyte ATGL can mitigate the adverse cardiac effects of chronic adrenergic stimulation and explored the underlying mechanisms. To do this, isoproterenol (ISO) was continuously administered to C57Bl/6N mice for 2 wk with or without an ATGL inhibitor (Atglistatin). We found that Atglistatin alleviated ISO-induced cardiac remodeling and reduced ISO-induced upregulation of galectin-3, a marker of activated macrophages and a potent inducer of fibrosis, in white adipose tissue (WAT), heart, and the circulation. To test whether the beneficial effects of Atglistatin occur via inhibition of adipocyte ATGL, adipocyte-specific ATGL knockout (atATGL-KO) mice were utilized for similar experiments. Subsequently, the same cardioprotective effects of atATGL-KO following ISO administration were observed. Furthermore, Atglistatin and atATGL-KO abolished ISO-induced galectin-3 secretion from excised WAT. We further demonstrated that activation of cardiac fibroblasts by the conditioned media of ISO-stimulated WAT is galectin-3-dependent. In conclusion, the inhibition of adipocyte ATGL ameliorated ISO-induced cardiac remodeling possibly by reducing galectin-3 secretion from adipose tissue. Thus, inhibition of adipocyte ATGL might be a potential target to prevent some of the adverse effects of chronic excess adrenergic drive.NEW & NOTEWORTHY The reduction of lipolysis by adipocyte ATGL inhibition ameliorates cardiac remodeling induced by chronic ß-adrenergic stimulation likely via reducing galectin-3 secretion from adipose tissue. Our findings highlight that suppressing lipolysis in adipocytes may be a potential therapeutic target for patients with heart failure whose sympathetic nervous system is activated. Furthermore, galectin-3 might be involved in the mechanisms by which excessive lipolysis in adipose tissues influences remote cardiac pathologies and thus warrants further investigation.


Subject(s)
Adipose Tissue, White/drug effects , Anti-Inflammatory Agents/pharmacology , Enzyme Inhibitors/pharmacology , Heart Diseases/prevention & control , Inflammation Mediators/metabolism , Isoproterenol , Lipase/antagonists & inhibitors , Phenylurea Compounds/pharmacology , Ventricular Remodeling/drug effects , Adipose Tissue, White/enzymology , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Galectin 3/metabolism , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/physiopathology , Lipase/metabolism , Lipolysis/drug effects , Male , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Paracrine Communication , Signal Transduction
5.
Front Immunol ; 11: 1953, 2020.
Article in English | MEDLINE | ID: mdl-32973799

ABSTRACT

Obesity is a major risk factor for the development of insulin resistance and type 2 diabetes. However, the mechanisms that trigger the underlying adipose tissues inflammation are not completely understood. Here, we show that the E3 ubiquitin ligase March1 controls the phenotypic and functional properties of CD8+ T cells in mice white adipose tissue. In a diet-induced obesity model, mice lacking March1 [March1 knockout (KO)] show increased insulin resistance compared to their WT counterparts. Also, in obese March1 KO mice, the proportions of effector/memory (Tem) and resident/memory (Trm) CD8+ T cells were higher in the visceral adipose tissue, but not in the spleen. The effect of March1 on insulin resistance and on the phenotype of adipose tissue CD8+ T cells was independent of major histocompatibility complex class II ubiquitination. Interestingly, we adoptively transferred either WT or March1 KO splenic CD8+ T cells into obese WT chimeras that had been reconstituted with Rag1-deficient bone marrow. We observed an enrichment of Tem and Trm cells and exacerbated insulin resistance in mice that received March1 KO CD8 T cells. Mechanistically, we found that March1 deficiency alters the metabolic activity of CD8+ T cells. Our results provide additional evidence of the involvement of CD8+ T cells in adipose tissue inflammation and suggest that March1 controls the metabolic reprogramming of these cells.


Subject(s)
Adipose Tissue, White/enzymology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Insulin Resistance , Obesity/enzymology , Ubiquitin-Protein Ligases/deficiency , Adipose Tissue, White/immunology , Adoptive Transfer , Animals , Blood Glucose/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/immunology , Phenotype , Spleen/enzymology , Spleen/immunology , Ubiquitin-Protein Ligases/genetics
6.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326642

ABSTRACT

Autophagy can remove excess or dysfunctional proteins and organelles to maintain cellular homeostasis. Browning of white adipose tissue increases the energy expenditure. Microtubules affinity regulated kinase 4 (Mark4) can regulate a variety of physiological processes. According to previous studies, we speculated that Mark4-autophagy-browning of white adipose tissue had certain linkages. Here, we established two autophagy models through serum starvation and rapamycin treatment and detected that the overexpression of Mark4 increased the expression of autophagy-related factors Beclin1, ATG7, and significantly decreased the autophagy substrate P62. Further tests showed that the overexpression of Mark4 promoted the conversion of autophagy marker protein LC3A to LC3B-II by activating the AMP-activated protein kinase (AMPK) pathway and inhibition of the AKT/mTOR signaling. Moreover, Mark4 decreased the expression of thermogenesis genes via promoting autophagy. These results indicated that Mark4 inhibited the browning of white adipose tissue via promoting autophagy.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Autophagy/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , 3T3 Cells , AMP-Activated Protein Kinase Kinases , Adipocytes/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/enzymology , Animals , Autophagy/drug effects , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Male , Mice , Microtubule-Associated Proteins/metabolism , Nutritive Value/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Stress, Physiological/physiology , TOR Serine-Threonine Kinases/metabolism , Thermogenesis , Up-Regulation
7.
Biochem J ; 477(8): 1373-1389, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32215608

ABSTRACT

We investigated acute effects of two allosteric protein kinase B (PKB) inhibitors, MK-2206 and Akti-1/2, on insulin-stimulated lipogenesis in rat epididymal adipocytes incubated with fructose as carbohydrate substrate. In parallel, the phosphorylation state of lipogenic enzymes in adipocytes and incubated epididymal fat pads was monitored by immunoblotting. Preincubation of rat epididymal adipocytes with PKB inhibitors dose-dependently inhibited the following: insulin-stimulated lipogenesis, increased PKB Ser473 phosphorylation, increased PKB activity and decreased acetyl-CoA carboxylase (ACC) Ser79 phosphorylation. In contrast, the effect of insulin to decrease the phosphorylation of pyruvate dehydrogenase (PDH) at Ser293 and Ser300 was not abolished by PKB inhibition. Insulin treatment also induced ATP-citrate lyase (ACL) Ser454 phosphorylation, but this effect was less sensitive to PKB inhibitors than ACC dephosphorylation by insulin. In incubated rat epididymal fat pads, Akti-1/2 treatment reversed insulin-induced ACC dephosphorylation, while ACL phosphorylation by insulin was maintained. ACL and ACC purified from white adipose tissue were poor substrates for PKBα in vitro. However, effects of wortmannin and torin, along with Akti-1/2 and MK-2206, on recognized PKB target phosphorylation by insulin were similar to their effects on insulin-induced ACL phosphorylation, suggesting that PKB could be the physiological kinase for ACL phosphorylation by insulin. In incubated epididymal fat pads from wild-type versus ACC1/2 S79A/S212A knockin mice, effects of insulin to increase lipogenesis from radioactive fructose or from radioactive acetate were reduced but not abolished. Together, the results support a key role for PKB in mediating insulin-stimulated lipogenesis by decreasing ACC phosphorylation, but not by decreasing PDH phosphorylation.


Subject(s)
Adipose Tissue, White/drug effects , Benzylamines/administration & dosage , Heterocyclic Compounds, 3-Ring/administration & dosage , Insulin/metabolism , Lipogenesis/drug effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quinoxalines/administration & dosage , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adipocytes/drug effects , Adipocytes/enzymology , Adipocytes/metabolism , Adipose Tissue, White/enzymology , Adipose Tissue, White/metabolism , Animals , Male , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
8.
Biomolecules ; 10(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-32131495

ABSTRACT

Biliverdin reductase (BVR) is an enzymatic and signaling protein that has multifaceted roles in physiological systems. Despite the wealth of knowledge about BVR, no data exist regarding its actions in adipocytes. Here, we generated an adipose-specific deletion of biliverdin reductase-A (BVRA) (BlvraFatKO) in mice to determine the function of BVRA in adipocytes and how it may impact adipose tissue expansion. The BlvraFatKO and littermate control (BlvraFlox) mice were placed on a high-fat diet (HFD) for 12 weeks. Body weights were measured weekly and body composition, fasting blood glucose and insulin levels were quantitated at the end of the 12 weeks. The data showed that the percent body fat and body weights did not differ between the groups; however, BlvraFatKO mice had significantly higher visceral fat as compared to the BlvraFlox. The loss of adipocyte BVRA decreased the mitochondrial number in white adipose tissue (WAT), and increased inflammation and adipocyte size, but this was not observed in brown adipose tissue (BAT). There were genes significantly reduced in WAT that induce the browning effect such as Ppara and Adrb3, indicating that BVRA improves mitochondria function and beige-type white adipocytes. The BlvraFatKO mice also had significantly higher fasting blood glucose levels and no changes in plasma insulin levels, which is indicative of decreased insulin signaling in WAT, as evidenced by reduced levels of phosphorylated AKT (pAKT) and Glut4 mRNA. These results demonstrate the essential role of BVRA in WAT in insulin signaling and adipocyte hypertrophy.


Subject(s)
Adipocytes, White/enzymology , Adipose Tissue, White/enzymology , Mitochondria/metabolism , Obesity/enzymology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Adipocytes, White/pathology , Adipose Tissue, White/pathology , Animals , Gene Knockout Techniques , Hypertrophy , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Obesity/genetics , Obesity/pathology , Oxidoreductases Acting on CH-CH Group Donors/genetics , PPAR alpha/genetics , PPAR alpha/metabolism , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism
9.
Am J Physiol Endocrinol Metab ; 318(3): E318-E329, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31961704

ABSTRACT

Browning of white adipose tissue (WAT) has been recognized as an important strategy for the treatment of obesity, insulin resistance, and diabetes. Enoyl coenzyme A hydratase 1 (ECH1) is a widely known enzyme involved in lipid metabolism. However, whether and how ECH1 is implicated in browning of WAT remain obscure. Adeno-associated, virus-mediated genetic engineering of ECH1 in adipose tissue was used in investigations in mouse models of obesity induced by a high-fat diet (HFD) or browning induced by cold exposure. Metabolic parameters showed that ECH1 overexpression decreased weight gain and improved insulin sensitivity and lipid profile after 8 wk of an HFD. Further work revealed that these changes were associated with enhanced energy expenditure and increased appearance of brown-like adipocytes in inguinal WAT, as verified by a remarkable increase in uncoupling protein 1 and thermogenic gene expression. In vitro, ECH1 induced brown fat-related gene expression in adipocytes differentiated from primary stromal vascular fractions, whereas knockdown of ECH1 reversed this effect. Mechanistically, ECH1 regulated the thermogenic program by inhibiting mammalian target of rapamycin signaling, which may partially explain the potential mechanism for ECH1 regulating adipose browning. In summary, ECH1 may participate in the pathology of obesity by regulating browning of WAT, which probably provides us with a new therapeutic strategy for combating obesity.


Subject(s)
Adipose Tissue, Brown/enzymology , Carbon-Carbon Double Bond Isomerases/genetics , Carbon-Carbon Double Bond Isomerases/metabolism , Genetic Therapy/methods , Metabolic Diseases/therapy , Obesity/therapy , Adipose Tissue, Brown/growth & development , Adipose Tissue, White/enzymology , Adipose Tissue, White/growth & development , Animals , Cold Temperature , Diet, High-Fat , Energy Metabolism , Genetic Engineering , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , TOR Serine-Threonine Kinases/metabolism , Thermogenesis , Weight Gain
10.
Am J Physiol Cell Physiol ; 318(1): C137-C149, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31721616

ABSTRACT

Reactive oxygen species (ROS) are important signaling molecules mediating the exercise-induced adaptations in skeletal muscle. Acute exercise also drives the expression of genes involved in reesterification and glyceroneogenesis in white adipose tissue (WAT), but whether ROS play any role in this effect has not been explored. We speculated that exercise-induced ROS would regulate acute exercise-induced responses in WAT. To address this question, we utilized various models to alter redox signaling in WAT. We examined basal and exercise-induced gene expression in a genetically modified mouse model of reduced mitochondrial ROS emission [mitochondrial catalase overexpression (MCAT)]. Additionally, H2O2, various antioxidants, and the ß3-adrenergic receptor agonist CL316243 were used to assess gene expression in white adipose tissue culture. MCAT mice have reduced ROS emission from WAT, enlarged WAT depots and adipocytes, and greater pyruvate dehydrogenase kinase-4 (Pdk4) gene expression. In WAT culture, H2O2 reduced glyceroneogenic gene expression. In wild-type mice, acute exercise induced dramatic but transient increases in Pdk4 and phosphoenolpyruvate carboxykinase (Pck1) mRNA in both subcutaneous inguinal WAT and epididymal WAT depots, which was almost completely absent in MCAT mice. Furthermore, the induction of Pdk4 and Pck1 in WAT culture by CL316243 was markedly reduced in the presence of antioxidants N-acetyl-cysteine or vitamin E. Genetic and nutritional approaches that attenuate redox signaling prevent exercise- and ß-agonist-induced gene expression within WAT. Combined, these data suggest that ROS represent important mediators of gene expression within WAT.


Subject(s)
Adipocytes/enzymology , Adipose Tissue, White/enzymology , Energy Metabolism , Mitochondria/enzymology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Reactive Oxygen Species/metabolism , Adipocytes/drug effects , Adipogenesis , Adipose Tissue, White/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Antioxidants , Catalase/genetics , Catalase/metabolism , Energy Metabolism/drug effects , Gene Expression Regulation, Enzymologic , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Oxidants/pharmacology , Oxidation-Reduction , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Physical Exertion , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Signal Transduction , Time Factors , Tissue Culture Techniques
11.
Nutrients ; 11(10)2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31618980

ABSTRACT

Gynostemma pentaphyllum is widely used in Asia as a herbal medicine to treat type 2 diabetes, dyslipidemia, and inflammation. Here, we investigated the anti-obesity effect and underlying mechanism of G. pentaphyllum extract (GPE) enriched in gypenoside L, gypenoside LI, and ginsenoside Rg3 and obtained using a novel extraction method. Five-week-old male C57BL/6N mice were fed a control diet (CD), high-fat diet (HFD), HFD + 100 mg/kg body weight (BW)/day GPE (GPE 100), HFD + 300 mg/kg BW/day GPE (GPE 300), or HFD + 30 mg/kg BW/day Orlistat (Orlistat 30) for 8 weeks. The HFD-fed mice showed significant increases in body weight, fat mass, white adipose tissue, and adipocyte hypertrophy compared to the CD group; but GPE inhibited those increases. GPE reduced serum levels of triglyceride, total cholesterol, and LDL-cholesterol, without affecting HDL-cholesterol. GPE significantly increased AMPK activation and suppressed adipogenesis by decreasing the mRNA expression of CCAAT/enhancer binding protein-α (C/EBPα), peroxisome proliferator-activated receptor-γ (PPARγ), sterol regulatory element-binding protein-1c (SREBP1c), PPARγ coactivator-1α, fatty acid synthase (FAS), adipocyte protein 2 (AP2), and sirtuin 1 (SIRT1) and by increasing that of carnitine palmitoyltransferase (CPT1) and hormone- sensitive lipase (HSL). This study demonstrated the ameliorative effect of GPE on obesity and elucidated the underlying molecular mechanism.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue, White/drug effects , Anti-Obesity Agents/pharmacology , Diet, High-Fat , Gynostemma/chemistry , Obesity/prevention & control , Plant Extracts/pharmacology , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, White/enzymology , Adipose Tissue, White/physiopathology , Adiposity/drug effects , Animals , Anti-Obesity Agents/isolation & purification , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Disease Models, Animal , Lipids/blood , Male , Mice, Inbred C57BL , Obesity/blood , Obesity/enzymology , Obesity/physiopathology , Oxidation-Reduction , Plant Extracts/isolation & purification , Signal Transduction , Up-Regulation , Weight Gain/drug effects
12.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1402-1409, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30776415

ABSTRACT

The nuclear receptor liver X receptor (LXR) impacts on cholesterol metabolism as well as hepatic lipogenesis via transcriptional regulation. It is proposed that inhibition of the protein arginine methyltransferase 3 (PRMT3) uncouples these two transcriptional pathways in vivo by acting as a specific lipogenic coactivator of LXR. Here we validated the hypothesis that treatment with the allosteric PRMT3 inhibitor SGC707 will diminish the hepatic steatosis extent, while leaving global cholesterol metabolism, important in cholesterol-driven pathologies like atherosclerosis, untouched. For this purpose, 12-week old hyperlipidemic apolipoprotein E knockout mice were fed a Western-type diet for six weeks to induce both hepatic steatosis and atherosclerosis. The mice received 3 intraperitoneal injections with SGC707 or solvent control per week. Mice chronically treated with SGC707 developed less severe hepatic steatosis as exemplified by the 51% reduced (P < 0.05) liver triglyceride levels. In contrast, the extent of in vivo macrophage foam cell formation and aortic root atherosclerosis was not affected by SGC707 treatment. Interestingly, SGC707-treated mice gained 94% less body weight (P < 0.05), which was paralleled by changes in white adipose tissue morphology, i.e. reduction in adipocyte size and browning. In conclusion, we have shown that through PRMT3 inhibitor treatment specific functions of LXR involved in respectively the development of fatty liver disease and atherosclerosis can be uncoupled, resulting in an overall diminished hepatic steatosis extent without a negative impact on atherosclerosis susceptibility. As such, our studies highlight that PRMT3 inhibition may constitute a novel therapeutic approach to limit the development of fatty liver disease in humans.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Enzyme Inhibitors/pharmacology , Fatty Liver/prevention & control , Isoquinolines/pharmacology , Protein-Arginine N-Methyltransferases/genetics , Adipose Tissue, White/drug effects , Adipose Tissue, White/enzymology , Adipose Tissue, White/pathology , Allosteric Regulation/drug effects , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/enzymology , Atherosclerosis/etiology , Atherosclerosis/pathology , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Susceptibility , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/pathology , Foam Cells/drug effects , Foam Cells/metabolism , Foam Cells/pathology , Gene Expression Regulation , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Mice , Mice, Knockout, ApoE , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction , Triglycerides/metabolism
13.
Int J Obes (Lond) ; 43(12): 2381-2393, 2019 12.
Article in English | MEDLINE | ID: mdl-30622312

ABSTRACT

OBJECTIVE: The lactation-suckling period is critical for white adipose tissue (WAT) development. Early postnatal nutrition influences later obesity risk but underlying mechanisms remain elusive. Here, we tested whether altered postnatal nutrition specifically during suckling impacts epigenetic regulation of key metabolic genes in WAT and alter long-term adiposity set point. METHODS: We analyzed the effects of maternal high-fat (HF) feeding in rats exclusively during lactation-suckling on breast milk composition and its impact on male offspring visceral epidydimal (eWAT) and subcutaneous inguinal (iWAT) depots during suckling and in adulthood. RESULTS: Maternal HF feeding during lactation had no effect on mothers' body weight (BW) or global breast milk composition, but induced qualitative changes in breast milk fatty acid (FA) composition (high n-6/n-3 polyunsaturated FA ratio and low medium-chain FA content). During suckling, HF neonates showed increased BW and mass of both eWAT and iWAT depot but only eWAT displayed an enhanced adipogenic transcriptional signature. In adulthood, HF offspring were predisposed to weight gain and showed increased hyperplastic growth only in eWAT. This specific eWAT expansion was associated with increased expression and activity of stearoyl-CoA desaturase-1 (SCD1), a key enzyme of FA metabolism. SCD1 converts saturated FAs, e.g. palmitate and stearate, to monounsaturated FAs, palmitoleate and oleate, which are the predominant substrates for triglyceride synthesis. Scd1 upregulation in eWAT was associated with reduced DNA methylation in Scd1 promoter surrounding a PPARγ-binding region. Conversely, changes in SCD1 levels and methylation were not observed in iWAT, coherent with a depot-specific programming. CONCLUSIONS: Our data reveal that maternal HF feeding during suckling programs long-term eWAT expansion in part by SCD1 epigenetic reprogramming. This programming events occurred with drastic changes in breast milk FA composition, suggesting that dietary FAs are key metabolic programming factors in the early postnatal period.


Subject(s)
Adipose Tissue, White , Diet, High-Fat , Epigenesis, Genetic/genetics , Lactation/genetics , Stearoyl-CoA Desaturase , Adipose Tissue, White/chemistry , Adipose Tissue, White/enzymology , Adipose Tissue, White/metabolism , Animals , Animals, Newborn , Body Weight/genetics , Female , Intra-Abdominal Fat/chemistry , Intra-Abdominal Fat/enzymology , Intra-Abdominal Fat/metabolism , Male , Milk/chemistry , Rats, Wistar , Stearoyl-CoA Desaturase/analysis , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
14.
Methods Mol Biol ; 1896: 31-38, 2019.
Article in English | MEDLINE | ID: mdl-30474837

ABSTRACT

Senescence-associated ß-galactosidase (hereafter SA-ß-gal) staining has now been employed for more than 20 years to identify the presence of senescent cells (Dimri et al., Proc Natl Acad Sci U S A 92:9363-9367, 1995). These cells, characterized by a permanent cell-cycle arrest (Hayflick and Moorhead, Exp Cell Res 25:585-621, 1961) and the production of a distinct secretory phenotype of cytokines, chemokines, and proteases (Coppe et al., PLoS Biol 6:2853-2868, 2008), have received much attention in recent years for their impacts on diverse biological processes. Here we describe a method to identify and quantify the specific cells that become senescent in vivo using transmission electron microscopy after SA-ß-gal staining that can be used in countless scenarios.


Subject(s)
Adipose Tissue, White/enzymology , Atherosclerosis/enzymology , Cellular Senescence , Kidney Tubules/enzymology , Pericardium/enzymology , beta-Galactosidase/metabolism , Adipose Tissue, White/cytology , Animals , Atherosclerosis/pathology , Cells, Cultured , Kidney Tubules/cytology , Mice , Pericardium/cytology
15.
FASEB J ; 33(1): 1428-1439, 2019 01.
Article in English | MEDLINE | ID: mdl-30133327

ABSTRACT

It is widely accepted that chronic stress may alter the homeostatic mechanisms of body weight control. In this study, we followed the metabolic changes occurring in mice when chronic stress caused by psychosocial defeat (CPD) is associated with ad libitum exposure to a palatable high-fat diet (HFD). In this model, CPD mice consumed more HFD than unstressed (Un) mice without gaining body weight. We focused on metabolic processes involved in weight control, such as de novo lipogenesis (DNL), fatty acid ß-oxidation (FAO), and thermogenesis. The activity and expression of DNL enzymes were reduced in the liver and white adipose tissue of mice consuming the HFD. Such effects were particularly evident in stressed mice. In both CPD and Un mice, HFD consumption increased the hepatic expression of the mitochondrial FAO enzyme carnitine palmitoyltransferase-1. In the liver of mice consuming the HFD, stress exposure prevented accumulation of triacylglycerols; however, accumulation of triacylglycerols was observed in Un mice under the same dietary regimen. In brown adipose tissue, stress increased the expression of uncoupling protein-1, which is involved in energy dissipation, both in HFD and control diet-fed mice. We consider increased FAO and energy dissipation responsible for the antiobesity effect seen in CPD/HFD mice. However, CPD associated with HFD induced hepatic oxidative stress.-Giudetti, A. M., Testini, M., Vergara, D., Priore, P., Damiano, F., Gallelli, C. A., Romano, A., Villani, R., Cassano, T., Siculella, L., Gnoni, G. V., Moles, A., Coccurello, R., Gaetani, S. Chronic psychosocial defeat differently affects lipid metabolism in liver and white adipose tissue and induces hepatic oxidative stress in mice fed a high-fat diet.


Subject(s)
Adipose Tissue, White/metabolism , Diet, High-Fat , Lipid Metabolism , Liver/metabolism , Oxidative Stress , Stress, Psychological , Acetyl-CoA Carboxylase/metabolism , Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/enzymology , Animals , Body Weight , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Disease Models, Animal , Energy Intake , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Glutathione/metabolism , Liver/enzymology , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , Uncoupling Protein 1/metabolism
16.
Cell Rep ; 24(4): 809-814, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30044978

ABSTRACT

Previous studies using genetic mouse models have implicated COX-2 in the browning of white adipose tissues (WATs) in mice during cold exposure. However, COX-2 is important during development, and conventional knockouts (KOs) exhibit many defects, conditioned by genetic background. Similarly, the physiological relevance of transgenic overexpression of COX-2 is questionable. In the present study, we utilized mice in which COX-2 was deleted postnatally, bypassing the consequences of enzyme deficiency during development. Despite activation of thermogenesis and browning of inguinal WAT, cold exposure failed to increase COX-2 expression in the adipose tissues of mice with different genetic backgrounds, and the body temperature response to cold was unaltered in postnatal global COX-2 KOs. Selective disruption of COX-2 in adipose tissues also failed detectably to impact systemic prostaglandin biosynthesis. Browning of inguinal WATs induced by exposure to cold is independent of adipose tissue COX-2.


Subject(s)
Adipose Tissue, Brown/enzymology , Adipose Tissue, White/enzymology , Cyclooxygenase 2/metabolism , Animals , Cold Temperature , Mice , Thermogenesis
17.
Nature ; 560(7716): 102-106, 2018 08.
Article in English | MEDLINE | ID: mdl-30022159

ABSTRACT

Thermogenesis by brown and beige adipose tissue, which requires activation by external stimuli, can counter metabolic disease1. Thermogenic respiration is initiated by adipocyte lipolysis through cyclic AMP-protein kinase A signalling; this pathway has been subject to longstanding clinical investigation2-4. Here we apply a comparative metabolomics approach and identify an independent metabolic pathway that controls acute activation of adipose tissue thermogenesis in vivo. We show that substantial and selective accumulation of the tricarboxylic acid cycle intermediate succinate is a metabolic signature of adipose tissue thermogenesis upon activation by exposure to cold. Succinate accumulation occurs independently of adrenergic signalling, and is sufficient to elevate thermogenic respiration in brown adipocytes. Selective accumulation of succinate may be driven by a capacity of brown adipocytes to sequester elevated circulating succinate. Furthermore, brown adipose tissue thermogenesis can be initiated by systemic administration of succinate in mice. Succinate from the extracellular milieu is rapidly metabolized by brown adipocytes, and its oxidation by succinate dehydrogenase is required for activation of thermogenesis. We identify a mechanism whereby succinate dehydrogenase-mediated oxidation of succinate initiates production of reactive oxygen species, and drives thermogenic respiration, whereas inhibition of succinate dehydrogenase supresses thermogenesis. Finally, we show that pharmacological elevation of circulating succinate drives UCP1-dependent thermogenesis by brown adipose tissue in vivo, which stimulates robust protection against diet-induced obesity and improves glucose tolerance. These findings reveal an unexpected mechanism for control of thermogenesis, using succinate as a systemically-derived thermogenic molecule.


Subject(s)
Adipose Tissue, Brown/metabolism , Succinic Acid/metabolism , Thermogenesis/physiology , Adipocytes/drug effects , Adipocytes/enzymology , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/enzymology , Adipose Tissue, White/cytology , Adipose Tissue, White/drug effects , Adipose Tissue, White/enzymology , Adipose Tissue, White/metabolism , Animals , Female , Male , Metabolomics , Mice , Obesity/metabolism , Obesity/prevention & control , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/metabolism , Succinic Acid/pharmacology , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism
18.
Nutrients ; 10(7)2018 Jun 27.
Article in English | MEDLINE | ID: mdl-29954059

ABSTRACT

Ginsenosides Rg1 is one of the major pharmacologically active saponins in ginseng, which as an antioxidant reduces oxidative damage in the liver and can also be used to prevent cardiovascular diseases and diabetes. However, there is no research targeting the effect of lipid metabolism in high-fat diet (HFD)-induced mice. In this study, we evaluated the anti-obesity effects of Rg1 in 3T3-L1 adipocyte cells and HFD-induced obese C57BL/6J mice. Administration of Rg1 to HFD-induced obese mice significantly decreased body weight, total cholesterol, and total triglyceride levels. In addition to effects in 3T3-L1 cells, Rg1 reduced the accumulation of lipid droplets in a dose-dependent manner. Furthermore, Rg1 exhibits an anti-adipogenic effect via regulation of the expression of the transcriptional factors and lipid metabolism-related genes in vivo and in vitro. We observed that Rg1 administration significantly increased the phosphorylation level of AMP-activated protein kinase (AMPK) in both epididymal white adipose tissue and 3T3-L1 cells. These results indicated that Rg1 works both in an anti-adipogenic and anti-obesity manner through inducing AMPK activation, inhibiting lipogenesis, and decreasing intracellular lipid content, adipocyte size, and adipose weight.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adipogenesis/drug effects , Adipose Tissue, White/drug effects , Anti-Obesity Agents/pharmacology , Diet, High-Fat , Ginsenosides/pharmacology , Lipogenesis/drug effects , Obesity/drug therapy , 3T3-L1 Cells , Adipocytes/enzymology , Adipocytes/pathology , Adipogenesis/genetics , Adipose Tissue, White/enzymology , Adipose Tissue, White/pathology , Adipose Tissue, White/physiopathology , Adiposity/drug effects , Animals , Biomarkers/blood , Cholesterol/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Lipid Droplets/drug effects , Lipid Droplets/enzymology , Lipogenesis/genetics , Male , Mice , Mice, Inbred C57BL , Obesity/enzymology , Obesity/pathology , Obesity/physiopathology , Phosphorylation , Signal Transduction/drug effects , Time Factors , Triglycerides/blood , Weight Loss/drug effects
19.
Toxicol Sci ; 164(1): 72-84, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29617909

ABSTRACT

Globally, approximately 10%-25% of women smoke during pregnancy. Since nicotine is highly addictive, women may use nicotine-containing products like nicotine replacement therapies for smoking cessation, but the long-term consequences of early life exposure to nicotine remain poorly defined. Our laboratory has previously demonstrated that maternal nicotine exposed (MNE) rat offspring exhibit hypertriglyceridemia due to increased hepatic de novo lipogenesis. Hypertriglyceridemia may also be attributed to impaired white adipose tissue (WAT) lipid storage; however, the effects of MNE on WAT are not completely understood. We hypothesize that nicotine-induced alterations in adipose function (eg, lipid storage) underlie dyslipidemia in MNE adults. Female 6-month-old rats exposed to nicotine during gestation and lactation exhibited significantly decreased visceral adipocyte cell area by 40%, attributed, in part, to a 3-fold increase in adipose triglyceride lipase (ATGL) protein expression compared with vehicle. Given ATGL has antioxidant properties and in utero nicotine exposure promotes oxidative stress in various tissues, we next investigated if there was evidence of increased oxidative stress in MNE WAT. At both 3 weeks and 6 months, MNE offspring expressed 37%-48% higher protein levels of superoxide dismutase-1 and -2 in WAT. Since oxidative stress can induce inflammation, we examined the inflammatory profile of WAT and found increased expression of cytokines (interleukin-1ß, tumor necrosis factor α, and interleukin-6) by 44%-61% at 6 months. Collectively, this suggests that the expression of WAT ATGL may be induced to counter MNE-induced oxidative stress and inflammation. However, higher levels of ATGL would further promote lipolysis in WAT, culminating in impaired lipid storage and long-term dyslipidemia.


Subject(s)
Adipose Tissue, White/drug effects , Antioxidants/metabolism , Lipase/genetics , Maternal Exposure/adverse effects , Nicotine/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Adipocytes, White/drug effects , Adipocytes, White/enzymology , Adipose Tissue, White/embryology , Adipose Tissue, White/enzymology , Adipose Tissue, White/growth & development , Animals , Escherichia coli Proteins/drug effects , Female , Lipogenesis/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/enzymology , Prenatal Exposure Delayed Effects/genetics , Rats, Wistar
20.
Biomed Pharmacother ; 103: 228-233, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29655163

ABSTRACT

Phosphofructokinase (PFK) is a key regulatory enzyme of glycolysis, being considered the pacemaker of this pathway. In mammals, this enzyme exists as three different isoforms, PFKM, PFKL and PFKP, presenting different regulatory and catalytic properties. The expression of these isoforms is tissue-specific and vary according to the cell differentiation and signalization. Although it is known that the expression of the different PFK isoforms directly affects cell function, the information regarding the regulation of PFK isoforms expression is scarce. In the present work, we evaluate the role of insulin signalization on the expression of three PFK isoforms on skeletal muscle, liver, and epididymal white adipose tissue (eWAT) of mice. For this, Swiss mice were treated with streptozotocin (STZ) to disrupt pancreatic ß-cells and, thus, insulin production. Control group were treated with citrate buffer (STZ vehicle). These groups were then treated with insulin or saline twice a day for ten consecutive days when animals were euthanized and tissues used for the evaluation of PFK isoforms expression by quantitative PCR (qPCR). Our results revealed that the lack of insulin significantly impacted the expression of PFKL, presenting mild effects on PFKM and no effects on PFKP. The decrease of PFKL and PFKM mRNA levels observed on the group treated with STZ was reversed by the treatment with insulin. In conclusion, insulin, the most known regulator of glucose consumption, specifically regulates the expression of PFKL and PFKM, which impact the regulation of glycolysis in the cell.


Subject(s)
Insulin/pharmacology , Liver/enzymology , Muscle, Skeletal/enzymology , Phosphofructokinase-1/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/enzymology , Animals , Isoenzymes/metabolism , Liver/drug effects , Male , Mice , Muscle, Skeletal/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL