Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.272
Filtrer
1.
Cell Metab ; 36(7): 1534-1549.e7, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38878772

RÉSUMÉ

Tirzepatide, a glucose-dependent insulinotropic polypeptide/glucagon-like peptide 1 receptor (GIPR/GLP-1R) agonist, has, in clinical trials, demonstrated greater reductions in glucose, body weight, and triglyceride levels compared with selective GLP-1R agonists in people with type 2 diabetes (T2D). However, cellular mechanisms by which GIPR agonism may contribute to these improved efficacy outcomes have not been fully defined. Using human adipocyte and mouse models, we investigated how long-acting GIPR agonists regulate fasted and fed adipocyte functions. In functional assays, GIPR agonism enhanced insulin signaling, augmented glucose uptake, and increased the conversion of glucose to glycerol in a cooperative manner with insulin; however, in the absence of insulin, GIPR agonists increased lipolysis. In diet-induced obese mice treated with a long-acting GIPR agonist, circulating triglyceride levels were reduced during oral lipid challenge, and lipoprotein-derived fatty acid uptake into adipose tissue was increased. Our findings support a model for long-acting GIPR agonists to modulate both fasted and fed adipose tissue function differentially by cooperating with insulin to augment glucose and lipid clearance in the fed state while enhancing lipid release when insulin levels are reduced in the fasted state.


Sujet(s)
Adipocytes , Souris de lignée C57BL , Récepteur hormone gastrointestinale , Animaux , Récepteur hormone gastrointestinale/métabolisme , Récepteur hormone gastrointestinale/agonistes , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Humains , Souris , Mâle , Insuline/métabolisme , Glucose/métabolisme , Lipolyse/effets des médicaments et des substances chimiques , Triglycéride/métabolisme , Peptide gastrointestinal/métabolisme , Peptide gastrointestinal/pharmacologie , Obésité/métabolisme , Obésité/traitement médicamenteux , Nutriments/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur du peptide-1 similaire au glucagon/métabolisme , Récepteur du peptide-1 similaire au glucagon/agonistes , Récepteur du peptide-2 similaire au glucagon
2.
Int J Mol Sci ; 25(9)2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38732183

RÉSUMÉ

The impact of microplastics (MPs) on the metabolic functions of the liver is currently unclear and not completely understood. To investigate the effects of the administration of MPs on the hepatic metabolism of normal and obese mice, alterations in the lipid, glucose (Glu), and amino acid regulation pathways were analyzed in the liver and adipose tissues of C57BL/6Korl (wild type, WT) or C57BL/6-Lepem1hwl/Korl mice (leptin knockout, Lep KO) orally administered polystyrene (PS) MPs for 9 weeks. Significant alterations in the lipid accumulation, adipogenesis, lipogenesis, and lipolysis pathways were detected in the liver tissue of MP-treated WT and Lep KO mice compared to the vehicle-treated group. These alterations in their liver tissues were accompanied by an upregulation of the serum lipid profile, as well as alterations in the adipogenesis, lipogenesis, and lipolysis pathways in the adipose tissues of MP-treated WT and Lep KO mice. Specifically, the level of leptin was increased in the adipose tissues of MP-treated WT mice without any change in their food intake. Also, MP-induced disruptions in the glycogenolysis, Glu transporter type 4 (GLUT4)-5' AMP-activated protein kinase (AMPK) signaling pathway, levels of lipid intermediates, and the insulin resistance of the liver tissues of WT and Lep KO mice were observed. Furthermore, the levels of seven endogenous metabolites were remarkably changed in the serum of WT and Lep KO mice after MP administrations. Finally, the impact of the MP administration observed in both types of mice was further verified in differentiated 3T3-L1 adipocytes and HepG2 cells. Thus, these results suggest that the oral administration of MPs for 9 weeks may be associated with the disruption of lipid, Glu, and amino acid metabolism in the liver tissue of obese WT and Lep KO mice.


Sujet(s)
Acides aminés , Glucose , Métabolisme lipidique , Foie , Souris de lignée C57BL , Souris knockout , Microplastiques , Polystyrènes , Animaux , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Souris , Glucose/métabolisme , Métabolisme lipidique/effets des médicaments et des substances chimiques , Acides aminés/métabolisme , Administration par voie orale , Leptine/métabolisme , Tissu adipeux/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques , Adipogenèse/effets des médicaments et des substances chimiques , Mâle , Lipogenèse/effets des médicaments et des substances chimiques , Obésité/métabolisme , Obésité/étiologie , Obésité/génétique , Humains , Lipolyse/effets des médicaments et des substances chimiques
3.
J Agric Food Chem ; 72(22): 12582-12595, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38788215

RÉSUMÉ

Renal tubular ectopic lipid deposition (ELD) plays a significant role in the development of chronic kidney disease, posing a great threat to human health. The present work aimed to explore the intervention effect and potential molecular mechanism of a purified tea polysaccharide (TPS3A) on renal tubular ELD. The results demonstrated that TPS3A effectively improved kidney function and slowed the progression of tubulointerstitial fibrosis in high-fat-diet (HFD)-exposed ApoE-/- mice. Additionally, TPS3A notably suppressed lipogenesis and enhanced lipolysis, as shown by the downregulation of lipogenesis markers (SREBP-1 and FAS) and the upregulation of lipolysis markers (HSL and ATGL), thereby reducing renal tubular ELD in HFD-fed ApoE-/- mice and palmitic-acid-stimulated HK-2 cells. The AMPK-SIRT1-FoxO1 axis is a core signal pathway in regulating lipid deposition. Consistently, TPS3A significantly increased the levels of phosphorylated-AMPK, SIRT1, and deacetylation of Ac-FoxO1. However, these effects of TPS3A on lipogenesis and lipolysis were abolished by AMPK siRNA, SIRT1 siRNA, and FoxO1 inhibitor, resulting in exacerbated lipid deposition. Taken together, TPS3A shows promise in ameliorating renal tubular ELD by inhibiting lipogenesis and promoting lipolysis through the AMPK-SIRT1-FoxO1 signaling pathway.


Sujet(s)
Alimentation riche en graisse , Lipogenèse , Lipolyse , Souris de lignée C57BL , Polyosides , Animaux , Lipogenèse/effets des médicaments et des substances chimiques , Souris , Lipolyse/effets des médicaments et des substances chimiques , Mâle , Alimentation riche en graisse/effets indésirables , Humains , Polyosides/pharmacologie , Polyosides/administration et posologie , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Tubules rénaux/métabolisme , Tubules rénaux/effets des médicaments et des substances chimiques , Camellia sinensis/composition chimique , AMP-Activated Protein Kinases/métabolisme , AMP-Activated Protein Kinases/génétique , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie , Thé/composition chimique , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique
4.
Biomed Pharmacother ; 176: 116799, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38805969

RÉSUMÉ

BACKGROUND: The overstoring of surplus calories in mature adipocytes causes obesity and abnormal metabolic activity. The anti-obesity effect of a Celosia cristata (CC) total flower extract was assessed in vitro, using 3T3-L1 pre-adipocytes and mouse adipose-derived stem cells (ADSCs), and in vivo, using high-fat diet (HFD)-treated C57BL/6 male mice. METHODS: CC extract was co-incubated during adipogenesis in both 3T3-L1 cells and ADSCs. After differentiation, lipid droplets were assessed by oil red O staining, adipogenesis and lipolytic factors were evaluated, and intracellular triglyceride and glycerol concentrations were analyzed. For in vivo experiments, histomorphological analysis, mRNA expression levels of adipogenic and lipolytic factors in adipose tissue, blood plasma analysis, metabolic profiles were investigated. RESULTS: CC treatment significantly prevented adipocyte differentiation and lipid droplet accumulation, reducing adipogenesis-related factors and increasing lipolysis-related factors. Consequently, the intracellular triacylglycerol content was diminished, whereas the glycerol concentration in the cell supernatant increased. Mice fed an HFD supplemented with the CC extract exhibited decreased HFD-induced weight gain with metabolic abnormalities such as intrahepatic lipid accumulation and adipocyte hypertrophy. Improved glucose utilization and insulin sensitivity were observed, accompanied by the amelioration of metabolic disturbances, including alterations in liver enzymes and lipid profiles, in CC-treated mice. Moreover, the CC extract helped restore the disrupted energy metabolism induced by the HFD, based on a metabolic animal monitoring system. CONCLUSION: This study suggests that CC total flower extract is a potential natural herbal supplement for the prevention and management of obesity.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Agents antiobésité , Celosia , Alimentation riche en graisse , Fleurs , Souris de lignée C57BL , Obésité , Extraits de plantes , Animaux , Extraits de plantes/pharmacologie , Extraits de plantes/isolement et purification , Mâle , Souris , Agents antiobésité/pharmacologie , Agents antiobésité/isolement et purification , Fleurs/composition chimique , Adipogenèse/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Obésité/métabolisme , Alimentation riche en graisse/effets indésirables , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Celosia/composition chimique , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/métabolisme , Métabolisme lipidique/effets des médicaments et des substances chimiques , Lipolyse/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques
5.
Sci Rep ; 14(1): 12430, 2024 05 30.
Article de Anglais | MEDLINE | ID: mdl-38816541

RÉSUMÉ

Dietary trans 10, cis 12-conjugated linoleic acid (t10c12-CLA) is a potential candidate in anti-obesity trials. A transgenic mouse was previously successfully established to determine the anti-obesity properties of t10c12-CLA in male mice that could produce endogenous t10c12-CLA. To test whether there is a different impact of t10c12-CLA on lipid metabolism in both sexes, this study investigated the adiposity and metabolic profiles of female Pai mice that exhibited a dose-dependent expression of foreign Pai gene and a shift of t10c12-CLA content in tested tissues. Compared to their gender-match wild-type littermates, Pai mice had no fat reduction but exhibited enhanced lipolysis and thermogenesis by phosphorylated hormone-sensitive lipase and up-regulating uncoupling proteins in brown adipose tissue. Simultaneously, Pai mice showed hepatic steatosis and hypertriglyceridemia by decreasing gene expression involved in lipid and glucose metabolism. Further investigations revealed that t10c10-CLA induced excessive prostaglandin E2, adrenaline, corticosterone, glucagon and inflammatory factors in a dose-dependent manner, resulting in less heat release and oxygen consumption in Pai mice. Moreover, fibroblast growth factor 21 overproduction only in monoallelic Pai/wt mice indicates that it was sensitive to low doses of t10c12-CLA. These results suggest that chronic t10c12-CLA has system-wide effects on female health via synergistic actions of various hormones.


Sujet(s)
Corticostérone , Dinoprostone , Épinéphrine , Facteurs de croissance fibroblastique , Glucagon , Acides linoléiques conjugués , Souris transgéniques , Animaux , Femelle , Facteurs de croissance fibroblastique/métabolisme , Facteurs de croissance fibroblastique/génétique , Souris , Acides linoléiques conjugués/pharmacologie , Acides linoléiques conjugués/métabolisme , Corticostérone/métabolisme , Dinoprostone/métabolisme , Glucagon/métabolisme , Épinéphrine/métabolisme , Thermogenèse/effets des médicaments et des substances chimiques , Thermogenèse/génétique , Mâle , Métabolisme lipidique/effets des médicaments et des substances chimiques , Tissu adipeux brun/métabolisme , Tissu adipeux brun/effets des médicaments et des substances chimiques , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Lipolyse/effets des médicaments et des substances chimiques , Hypertriglycéridémie/métabolisme , Hypertriglycéridémie/génétique , Adiposité/effets des médicaments et des substances chimiques
6.
J Pharm Pharmacol ; 76(7): 897-907, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38727186

RÉSUMÉ

OBJECTIVES: Bile acids (BAs), as signaling molecules to regulate metabolism, have received considerable attention. Genipin is an iridoid compound extracted from Fructus Gradeniae, which has been shown to relieve adiposity and metabolic syndrome. Here, we investigated the mechanism of genipin counteracting obesity and its relationship with BAs signals in diet-induced obese (DIO) rats. METHODS: The DIO rats were received intraperitoneal injections of genipin for 10 days. The body weight, visceral fat, lipid metabolism in the liver, thermogenic genes expressions in brown fat, BAs metabolism and signals, and key enzymes for BAs synthesis were determined. KEY FINDINGS: Genipin inhibited fat synthesis and promoted lipolysis in the liver, and upregulated thermogenic gene expressions in brown adipose tissue of DIO rats. Genipin increased bile flow rate and upregulated the expressions of aquaporin 8 and the transporters of BAs in liver. Furthermore, genipin changed BAs composition by promoting alternative pathways and inhibiting classical pathways for BAs synthesis and upregulated the expressions of bile acid receptors synchronously. CONCLUSIONS: These results suggest that genipin ameliorate obesity through BAs-mediated signaling pathways.


Sujet(s)
Acides et sels biliaires , Iridoïdes , Foie , Obésité , Rat Sprague-Dawley , Animaux , Obésité/traitement médicamenteux , Obésité/métabolisme , Iridoïdes/pharmacologie , Acides et sels biliaires/métabolisme , Mâle , Rats , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Bile/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Lipolyse/effets des médicaments et des substances chimiques , Graisse intra-abdominale/effets des médicaments et des substances chimiques , Graisse intra-abdominale/métabolisme
7.
Cell Rep ; 43(6): 114288, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38814782

RÉSUMÉ

Lipids have emerged as potent regulators of immune cell function. In the skin, adipocyte lipolysis increases the local pool of free fatty acids and is essential for coordinating early macrophage inflammation following injury. Here, we investigate G-protein-coupled receptor 84 (GPR84), a medium-chain fatty acid (MCFA) receptor, for its potential to propagate pro-inflammatory signaling after skin injury. GPR84 signaling was identified as a key component of regulating myeloid cell numbers and subsequent tissue repair through in vivo administration of a pharmacological antagonist and the MCFA decanoic acid. We found that impaired injury-induced dermal adipocyte lipolysis is a hallmark of diabetes, and lipidomic analysis demonstrated that MCFAs are significantly reduced in diabetic murine wounds. Furthermore, local administration of decanoic acid rescued myeloid cell numbers and tissue repair during diabetic wound healing. Thus, GPR84 is a readily targetable lipid signaling pathway for manipulating injury-induced tissue inflammation with beneficial effects on acute diabetic healing.


Sujet(s)
Diabète expérimental , Inflammation , Récepteurs couplés aux protéines G , Peau , Cicatrisation de plaie , Animaux , Mâle , Souris , Adipocytes/métabolisme , Acides capriques/pharmacologie , Diabète expérimental/métabolisme , Diabète expérimental/anatomopathologie , Inflammation/anatomopathologie , Inflammation/métabolisme , Lipolyse/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cellules myéloïdes/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Transduction du signal , Peau/anatomopathologie , Peau/métabolisme , Peau/traumatismes , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Femelle
8.
Mol Cell Endocrinol ; 588: 112225, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38570133

RÉSUMÉ

Although Liraglutide (Lira) increases serum irisin levels in type 2 diabetes mellitus (T2DM), it is unclear whether it induces expression of uncoupling protein 1 (UCP1) of adipocytes via promoting irisin secretion from skeletal muscle. Male T2DM rats were treated with 0.4 mg/kg/d Lira twice a day for 8 weeks, and the protein expression of phosphorylated AMP kinase (p-AMPK), phosphorylated acetyl-CoA carboxylase 1 (p-ACC1) and UCP1 in white adipose tissues were detected. Differentiated C2C12 cells were treated with palmitic acid (PA) and Lira to detect the secretion of irisin. Differentiated 3T3-L1 cells were treated with irisin, supernatant from Lira-treated C2C12 cells, Compound C or siAMPKα1, the triglyceride (TG) content and the related gene expression were measured. The transcriptome in irisin-treated differentiated 3T3-L1 cells was analyzed. Lira elevated serum irisin levels, decreased the adipocyte size and increased the protein expression of UCP1, p-AMPK and p-ACC1 in WAT. Moreover, it promoted the expression of PGC1α and FNDC5, the secretion of irisin in PA-treated differentiated C2C12 cells. The irisin and supernatant decreased TG synthesis and promoted the expression of browning- and lipolysis-related genes in differentiated 3T3-L1 cells. While Compound C and siAMPKα1 blocked AMPK activities and expression, irisin partly reversed the pathway. Finally, the transcriptome analysis indicated that differently expressed genes are mainly involved in browning and lipid metabolism. Overall, our findings showed that Lira modulated muscle-to-adipose signaling pathways in diabetes via irisin-mediated AMPKα/ACC1/UCP1/PPARα pathway. Our results suggest a new mechanism for the treatment of T2DM by Lira.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Fibronectines , Lipolyse , Liraglutide , Protéine-1 de découplage , Animaux , Fibronectines/métabolisme , Fibronectines/génétique , Souris , Protéine-1 de découplage/métabolisme , Protéine-1 de découplage/génétique , Mâle , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Lipolyse/effets des médicaments et des substances chimiques , Liraglutide/pharmacologie , Rats , Diabète expérimental/métabolisme , Diabète expérimental/traitement médicamenteux , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Rat Sprague-Dawley , Différenciation cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques
9.
Cell Rep Med ; 5(5): 101525, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38663398

RÉSUMÉ

Spinal cord injury (SCI) increases the risk of cardiometabolic disorders, including hypertension, dyslipidemia, and insulin resistance. Not only does SCI lead to pathological expansion of adipose tissue, but it also leads to ectopic lipid accumulation in organs integral to glucose and insulin metabolism. The pathophysiological changes that underlie adipose tissue dysfunction after SCI are unknown. Here, we find that SCI exacerbates lipolysis in epididymal white adipose tissue (eWAT). Whereas expression of the α2δ1 subunit of voltage-gated calcium channels increases in calcitonin gene-related peptide-positive dorsal root ganglia neurons that project to eWAT, conditional deletion of the gene encoding α2δ1 in these neurons normalizes eWAT lipolysis after SCI. Furthermore, α2δ1 pharmacological blockade through systemic administration of gabapentin also normalizes eWAT lipolysis after SCI, preventing ectopic lipid accumulation in the liver. Thus, our study provides insight into molecular causes of maladaptive sensory processing in eWAT, facilitating the development of strategies to reduce metabolic and cardiovascular complications after SCI.


Sujet(s)
Tissu adipeux blanc , Homéostasie , Lipolyse , Traumatismes de la moelle épinière , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/anatomopathologie , Traumatismes de la moelle épinière/physiopathologie , Animaux , Lipolyse/effets des médicaments et des substances chimiques , Mâle , Souris , Tissu adipeux blanc/métabolisme , Plasticité neuronale/effets des médicaments et des substances chimiques , Ganglions sensitifs des nerfs spinaux/métabolisme , Ganglions sensitifs des nerfs spinaux/anatomopathologie , Tissu adipeux/métabolisme , Souris de lignée C57BL , Neurones/métabolisme , Neurones/anatomopathologie , Peptide relié au gène de la calcitonine/métabolisme , Peptide relié au gène de la calcitonine/génétique
10.
Mol Cell Endocrinol ; 589: 112250, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38663485

RÉSUMÉ

The most common form of hypercortisolism is iatrogenic Cushing's syndrome. Lipodystrophy and metabolic disorders can result from the use of exogenous glucocorticoids (GC). Adipocytes play an important role in the production of circulating exosomal microRNAs, and knockdown of Dicer promotes lipodystrophy. The aim of this study is to investigate the effect of GCs on epididymal fat and to assess their influence on circulating microRNAs associated with fat turnover. The data indicate that despite the reduction in adipocyte volume due to increased lipolysis and apoptosis, there is no difference in tissue mass, suggesting that epididymal fat pad, related to animal size, is not affected by GC treatment. Although high concentrations of GC have no direct effect on epididymal microRNA-150-5p expression, GC can induce epididymal adipocyte uptake of microRNA-150-5p, which regulates transcription factor Ppar gamma during adipocyte maturation. In addition, GC treatment increased lipolysis and decreased glucose-derived lipid and glycerol incorporation. In conclusion, the similar control and GC epididymal fat mass results from increased dense fibrogenic tissue and decreased adipocyte volume induced by the lipolytic effect of GC. These findings demonstrate the complexity of epididymal fat. They also highlight how this disease alters fat distribution. This study is the first in a series published by our laboratory showing the detailed mechanism of adipocyte turnover in this disease.


Sujet(s)
Adipocytes , Épididyme , Glucocorticoïdes , Lipolyse , microARN , Mâle , Animaux , microARN/métabolisme , microARN/génétique , Épididyme/effets des médicaments et des substances chimiques , Épididyme/métabolisme , Épididyme/anatomopathologie , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Glucocorticoïdes/effets indésirables , Glucocorticoïdes/pharmacologie , Lipolyse/effets des médicaments et des substances chimiques , Souris , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique
11.
Mol Cell Endocrinol ; 589: 112249, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38604550

RÉSUMÉ

Using a three-dimensional (3-D) in vitro culture model, we report the dose dependent effect of 17ß-estradiol and testosterone on the adipogenic differentiation and maturation of human adipose derived stem cells (hASCs) obtained from female and male patients. Considering sexual dimorphism, we expected male and female adipocytes to respond differently to the sex steroids. Both male and female hASC spheroids were exposed to 100 nM and 500 nM of 17ß-estradiol and testosterone either at the beginning of the adipogenic maturation (Phase I) to discourage intracellular triglyceride accumulation or exposed after adipogenic maturation (Phase II) to reduce the intracellular triglyceride accumulation. The results show that 17ß-estradiol leads to a dose dependent reduction in intracellular triglyceride accumulation in female hASC spheroids compared to the both untreated and testosterone-treated cells. Affirming our hypothesis, 17ß-estradiol prevented intracellular triglyceride accumulation during Phase I, while it stimulated lipolysis during Phase II. PPAR-γ and adiponectin gene expression also reduced upon 17ß-estradiol treatment in female cells. Interestingly, 17ß-estradiol and testosterone had only a modest effect on the male hASC spheroids. Collectively, our findings suggest that 17ß-estradiol can prevent fat accumulation in adipocytes during early and late stages of maturation in females.


Sujet(s)
Adipogenèse , Adiponectine , Oestradiol , Caractères sexuels , Testostérone , Humains , Adipogenèse/effets des médicaments et des substances chimiques , Mâle , Femelle , Oestradiol/pharmacologie , Testostérone/pharmacologie , Adiponectine/métabolisme , Triglycéride/métabolisme , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/cytologie , Cellules cultivées , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Adipocytes/cytologie , Cellules souches/métabolisme , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/cytologie , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques , Lipolyse/effets des médicaments et des substances chimiques
12.
J Pharm Pharmacol ; 76(7): 834-841, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38588466

RÉSUMÉ

OBJECTIVES: Madecassoside (MA) is a triterpene derived from Centella asiatica that has been recognized for its antioxidant and anti-inflammatory properties in various disease models. However, its direct impact on cultured white adipocytes and the underlying mechanisms, mainly through gene knockdown, have not been thoroughly explored. METHODS: Western blot analysis was utilized to assess the expression levels of various proteins, while oil red O staining was used to measure lipid deposition. The adipocyte shapes were confirmed using H&E staining. KEY FINDINGS: MA treatment enhanced browning and lipolysis in 3T3-L1 adipocytes and adipose tissue from experimental mice while suppressing lipogenesis. Furthermore, MA treatment increased the expression of PPARα and FGF21 in 3T3-L1 adipocytes as well as the secretion of FGF21 into the culture medium. Knockdown of PPARα or FGF21 using siRNA diminished the effects of MA on lipid metabolism in cultured adipocytes. CONCLUSIONS: These findings demonstrate that MA promotes thermogenic browning and lipolysis while inhibiting adipocyte lipogenesis, thus showing the potential for attenuating obesity. The study suggested that MA could be a viable therapeutic approach for treating obesity.


Sujet(s)
Cellules 3T3-L1 , Facteurs de croissance fibroblastique , Lipogenèse , Lipolyse , Obésité , Récepteur PPAR alpha , Triterpènes , Animaux , Souris , Lipolyse/effets des médicaments et des substances chimiques , Triterpènes/pharmacologie , Lipogenèse/effets des médicaments et des substances chimiques , Facteurs de croissance fibroblastique/métabolisme , Récepteur PPAR alpha/métabolisme , Obésité/métabolisme , Obésité/traitement médicamenteux , Mâle , Métabolisme lipidique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Thermogenèse/effets des médicaments et des substances chimiques , Adipocytes blancs/effets des médicaments et des substances chimiques , Adipocytes blancs/métabolisme
13.
J Med Food ; 27(6): 477-487, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38498802

RÉSUMÉ

Obesity is a threat to public health and effective new medications are required. Platycodonis Radix (PR) is a traditional medicinal/dietary plant with activities against obesity. Using mice given a diet rich in fat, the antiobesity components of PR were identified and their molecular mechanisms were clarified further in this investigation. Initially, the impacts of PR fractions on liver histology and biochemical markers were assessed. Subsequently, the degrees of lipogenic and lipolytic gene and protein expressions were determined. Oral administration of PR polysaccharides (PG) (0.80 g/kg body weight) improved liver function (alanine aminotransferase and aspartate aminotransferase) and its antioxidant activities (total superoxide dismutase, glutathione peroxidase, and malondialdehyde), as well as alleviated blood lipid (total cholesterol, total triglyceride, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol) values, inflammatory systemic (TNF-α and IL-1ß), and histological abnormalities within the liver. Furthermore, PG administration downregulated the expression for lipogenic genes (ACC and FAS) and upregulated the expression for the lipolytic gene (PPARα, LPL, CPT1, and HSL). Importantly, PG raised AMPK phosphorylation and decreased SREBP-1c protein synthesis. Thus, it is possible that PG stimulates the AMPK-LPL/HSL path (lipolytic route) plus the AMPK-ACC/PPARα-CPT1 path (associated to ß-oxidation of fatty acids), while inhibiting the AMPK/(SREBP-1c)-ACC/FAS path (lipogenic route). In summary, PG has the ability to regulate lipid metabolism, and it may be useful to pharmacologically activate AMPK with PG to prevent and cure obesity.


Sujet(s)
Agents antiobésité , Alimentation riche en graisse , Foie , Souris de lignée C57BL , Obésité , Extraits de plantes , Platycodon , Animaux , Alimentation riche en graisse/effets indésirables , Obésité/métabolisme , Obésité/traitement médicamenteux , Mâle , Agents antiobésité/pharmacologie , Agents antiobésité/administration et posologie , Souris , Platycodon/composition chimique , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie , Humains , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Racines de plante/composition chimique , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/génétique , Polyosides/pharmacologie , Polyosides/administration et posologie , Lipogenèse/effets des médicaments et des substances chimiques , Lipolyse/effets des médicaments et des substances chimiques , Triglycéride/métabolisme , Triglycéride/sang , Alanine transaminase/métabolisme , Alanine transaminase/sang
14.
Autophagy ; 20(6): 1383-1397, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38436206

RÉSUMÉ

BRAFV600E is the most prevalent mutation in thyroid cancer and correlates with poor prognosis and therapy resistance. Although selective inhibitors of BRAFV600E have been developed, more advanced tumors such as anaplastic thyroid carcinomas show a poor response in clinical trials. Therefore, the study of alternative survival mechanisms is needed. Since metabolic changes have been related to malignant progression, in this work we explore metabolic dependencies of thyroid tumor cells to exploit them therapeutically. Our results show that respiration of thyroid carcinoma cells is highly dependent on fatty acid oxidation and, in turn, fatty acid mitochondrial availability is regulated through macroautophagy/autophagy. Furthermore, we show that both lysosomal inhibition and the knockout of the essential autophagy gene, ATG7, lead to enhanced lipolysis; although this effect is not essential for survival of thyroid carcinoma cells. We also demonstrate that following inhibition of either autophagy or fatty acid oxidation, thyroid tumor cells compensate oxidative phosphorylation deficiency with an increase in glycolysis. In contrast to lipolysis induction, upon autophagy inhibition, glycolytic boost in autophagy-deficient cells is essential for survival and, importantly, correlates with a higher sensitivity to the BRAFV600E selective inhibitor, vemurafenib. In agreement, downregulation of the glycolytic pathway results in enhanced mitochondrial respiration and vemurafenib resistance. Our work provides new insights into the role of autophagy in thyroid cancer metabolism and supports mitochondrial targeting in combination with vemurafenib to eliminate BRAFV600E-positive thyroid carcinoma cells.Abbreviations: AMP: adenosine monophosphate; ATC: anaplastic thyroid carcinoma; ATG: autophagy related; ATP: adenosine triphosphate; BRAF: B-Raf proto-oncogene, serine/threonine kinase; Cas9: CRISPR-associated protein; CREB: cAMP responsive element binding protein; CRISPR: clustered regularly interspaced short palindromic repeats; 2DG: 2-deoxyglucose; FA: fatty acid; FAO: fatty acid oxidation; FASN: fatty acid synthase; FCCP: trifluoromethoxy carbonyl cyanide phenylhydrazone; LAMP1: lysosomal associated membrane protein 1; LIPE/HSL: lipase E, hormone sensitive type; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; OCR: oxygen consumption rate; OXPHOS: oxidative phosphorylation; PRKA/PKA: protein kinase cAMP-activated; PTC: papillary thyroid carcinoma; SREBF1/SREBP1: sterol regulatory element binding transcription factor 1.


Sujet(s)
Autophagie , Résistance aux médicaments antinéoplasiques , Mitochondries , Protéines proto-oncogènes B-raf , Tumeurs de la thyroïde , Humains , Autophagie/effets des médicaments et des substances chimiques , Autophagie/génétique , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes B-raf/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Acides gras/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Proto-oncogène Mas , Vémurafénib/pharmacologie , Lipolyse/effets des médicaments et des substances chimiques , Respiration cellulaire/effets des médicaments et des substances chimiques , Protéine-7 associée à l'autophagie/métabolisme , Protéine-7 associée à l'autophagie/génétique , Sulfonamides/pharmacologie , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Indoles/pharmacologie
15.
J Dairy Sci ; 107(7): 5104-5121, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38278290

RÉSUMÉ

Dairy cows with clinical ketosis (CK) exhibit excessive adipose tissue (AT) lipolysis and systemic inflammation. Lipolysis in cows can be induced by the canonical (hormonally induced) and inflammatory lipolytic pathways. Currently, the most common treatment for CK is oral propylene glycol (PG); however, PG does not reduce lipolysis or inflammation. Niacin (NIA) can reduce the activation of canonical lipolysis, whereas cyclooxygenase inhibitors such as flunixin meglumine (FM) can limit inflammation and inhibit the inflammatory lipolytic pathway. The objective of this study was to determine the effects of including NIA and FM in the standard PG treatment for postpartum CK on AT function. Multiparous Jersey cows (n = 18; 7.1 ± 3.8 DIM) were selected from a commercial dairy. Inclusion criteria were CK symptoms (lethargy, depressed appetite, and drop in milk yield) and high blood levels of BHB (≥1.2 mmol/L). Cows with CK were randomly assigned to one of 3 treatments: (1) PG: 310 g administered orally once per day for 5 d, (2) PG+NIA: 24 g administered orally once per day for 3 d, and (3) PG+NIA+FM: 1.1 mg/kg administered IV once per day for 3 d. Healthy control cows (HC; n = 6) matched by lactation and DIM (±2 d) were sampled. Subcutaneous AT explants were collected at d 0 and d 7 relative to enrollment. To assess AT insulin sensitivity, explants were treated with insulin (1 µL/L) during lipolysis stimulation with a ß-adrenergic receptor agonist (isoproterenol, 1 µM). Lipolysis was quantified by glycerol release in the media. Lipid mobilization and inflammatory gene networks were evaluated using quantitative PCR. Protein biomarkers of lipolysis, insulin signaling, and AT inflammation, including hormone-sensitive lipase, protein kinase B (Akt), and ERK1/2, were quantified by capillary immunoassays. Flow cytometry of AT cellular components was used to characterize macrophage inflammatory phenotypes. Statistical significance was determined by a nonparametric t-test when 2 groups (HC vs. CK) were analyzed and an ANOVA test with Tukey adjustment when 3 treatment groups (PG vs. PG+NIA vs. PG+NIA+FM) were evaluated. At d 0, AT from CK cows showed higher mRNA expression of lipolytic enzymes ABHD5, LIPE, and LPL, as well as increased phosphorylation of hormone-sensitive lipase compared with HC. At d 0, insulin reduced lipolysis by 41% ± 8% in AT from HC, but CK cows were unresponsive (-2.9 ± 4%). Adipose tissue from CK cows exhibited reduced Akt phosphorylation compared with HC. Cows with CK had increased AT expression of inflammatory gene markers, including CCL2, IL8, IL10, TLR4, and TNF, along with ERK1/2 phosphorylation. Adipose tissue from CK cows showed increased macrophage infiltration compared with HC. By d 7, AT from PG+NIA+FM cows had a more robust response to insulin, as evidenced by reduced glycerol release (36.5% ± 8% compared with PG at 26.9% ± 7% and PG+NIA at 7.4% ± 8%) and enhanced phosphorylation of Akt. By d 7, PG+NIA+FM cows presented lower inflammatory markers, including ERK1/2 phosphorylation, and reduced macrophage infiltration, compared with PG and PG+NIA. These data suggest that including NIA and FM in CK treatment improves AT insulin sensitivity and reduces AT inflammation and macrophage infiltration.


Sujet(s)
Tissu adipeux , Maladies des bovins , Cétose , Lipolyse , Animaux , Bovins , Lipolyse/effets des médicaments et des substances chimiques , Femelle , Tissu adipeux/métabolisme , Cétose/médecine vétérinaire , Cétose/traitement médicamenteux , Maladies des bovins/traitement médicamenteux , Maladies des bovins/métabolisme , Lactation , Acide nicotinique/pharmacologie , Acide nicotinique/usage thérapeutique , Période du postpartum
16.
Nature ; 625(7993): 175-180, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38093006

RÉSUMÉ

Oxytocin (OXT), a nine-amino-acid peptide produced in the hypothalamus and released by the posterior pituitary, has well-known actions in parturition, lactation and social behaviour1, and has become an intriguing therapeutic target for conditions such as autism and schizophrenia2. Exogenous OXT has also been shown to have effects on body weight, lipid levels and glucose homeostasis1,3, suggesting that it may also have therapeutic potential for metabolic disease1,4. It is unclear, however, whether endogenous OXT participates in metabolic homeostasis. Here we show that OXT is a critical regulator of adipose tissue lipolysis in both mice and humans. In addition, OXT serves to facilitate the ability of ß-adrenergic agonists to fully promote lipolysis. Most surprisingly, the relevant source of OXT in these metabolic actions is a previously unidentified subpopulation of tyrosine hydroxylase-positive sympathetic neurons. Our data reveal that OXT from the peripheral nervous system is an endogenous regulator of adipose and systemic metabolism.


Sujet(s)
Tissu adipeux , Lipolyse , Neurones , Ocytocine , Animaux , Humains , Souris , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/métabolisme , Agonistes bêta-adrénergiques/pharmacologie , Lipolyse/effets des médicaments et des substances chimiques , Neurones/métabolisme , Ocytocine/métabolisme , Ocytocine/pharmacologie , Tyrosine 3-monooxygenase/métabolisme
18.
J Clin Endocrinol Metab ; 108(3): 653-664, 2023 02 15.
Article de Anglais | MEDLINE | ID: mdl-36240323

RÉSUMÉ

CONTEXT: Exogenous ketone body administration lowers circulating glucose levels but the underlying mechanisms are uncertain. OBJECTIVE: We tested the hypothesis that administration of the ketone body ß-hydroxybutyrate (ßOHB) acutely increases insulin sensitivity via feedback suppression of circulating free fatty acid (FFA) levels. METHODS: In a randomized, single-blinded crossover design, 8 healthy men were studied twice with a growth hormone (GH) infusion to induce lipolysis in combination with infusion of either ßOHB or saline. Each study day comprised a basal period and a hyperinsulinemic-euglycemic clamp combined with a glucose tracer and adipose tissue and skeletal muscle biopsies. RESULTS: ßOHB administration profoundly suppressed FFA levels concomitantly with a significant increase in glucose disposal and energy expenditure. This was accompanied by a many-fold increase in skeletal muscle content of both ßOHB and its derivative acetoacetate. CONCLUSION: Our data unravel an insulin-sensitizing effect of ßOHB, which we suggest is mediated by concomitant suppression of lipolysis.


Sujet(s)
Hormone de croissance humaine , Insulinorésistance , Corps cétoniques , Humains , Mâle , Acide 3-hydroxy-butyrique/pharmacologie , Acide gras libre , Glucose , Technique du clamp glycémique , Hormone de croissance , Hormone de croissance humaine/pharmacologie , Insuline/pharmacologie , Insulinorésistance/physiologie , Corps cétoniques/pharmacologie , Corps cétoniques/usage thérapeutique , Lipolyse/effets des médicaments et des substances chimiques , Lipolyse/physiologie
19.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-35806278

RÉSUMÉ

Casein kinase 2 (CK2) is a ubiquitously expressed serine/threonine kinase and is upregulated in human obesity. CX-4945 (Silmitasertib) is a CK2 inhibitor with anti-cancerous and anti-adipogenic activities. However, the anti-adipogenic and pro-lipolytic effects and the mode of action of CX-4945 in (pre)adipocytes remain elusive. Here, we explored the effects of CX-4945 on adipogenesis and lipolysis in differentiating and differentiated 3T3-L1 cells, a murine preadipocyte cell line. CX-4945 at 15 µM strongly reduced lipid droplet (LD) accumulation and triglyceride (TG) content in differentiating 3T3-L1 cells, indicating the drug's anti-adipogenic effect. Mechanistically, CX-4945 reduced the expression levels of CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and perilipin A in differentiating 3T3-L1 cells. Strikingly, CX-4945 further increased the phosphorylation levels of cAMP-activated protein kinase (AMPK) and liver kinase B-1 (LKB-1) while decreasing the intracellular ATP content in differentiating 3T3-L1 cells. In differentiated 3T3-L1 cells, CX-4945 had abilities to stimulate glycerol release and elevate the phosphorylation levels of hormone-sensitive lipase (HSL), pointing to the drug's pro-lipolytic effect. In addition, CX-4945 induced the activation of extracellular signal-regulated kinase-1/2 (ERK-1/2), and PD98059, an inhibitor of ERK-1/2, attenuated the CX4945-induced glycerol release and HSL phosphorylation in differentiated 3T3-L1 cells, indicating the drug's ERK-1/2-dependent lipolysis. In summary, this investigation shows that CX-4945 has strong anti-adipogenic and pro-lipolytic effects on differentiating and differentiated 3T3-L1 cells, mediated by control of the expression and phosphorylation levels of CK2, C/EBP-α, PPAR-γ, FAS, ACC, perilipin A, AMPK, LKB-1, ERK-1/2, and HSL.


Sujet(s)
Adipogenèse , Casein Kinase II , Naphtyridines , Phénazines , Cellules 3T3-L1 , AMP-Activated Protein Kinases/métabolisme , Animaux , Protéine alpha liant les séquences stimulatrices de type CCAAT/métabolisme , Casein Kinase II/antagonistes et inhibiteurs , Casein Kinase II/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cyclic AMP-Dependent Protein Kinases/métabolisme , Glycérol/pharmacologie , Humains , Lipolyse/effets des médicaments et des substances chimiques , Souris , Naphtyridines/pharmacologie , Récepteur PPAR gamma/métabolisme , Périlipine-1/métabolisme , Phénazines/pharmacologie , Sterol Esterase/métabolisme
20.
Int J Mol Sci ; 23(9)2022 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-35563078

RÉSUMÉ

Oncostatin M (OSM) is an immune cell-derived cytokine that is upregulated in adipose tissue in obesity. Upon binding its receptor (OSMR), OSM induces the phosphorylation of the p66 subunit of Src homology 2 domain-containing transforming protein 1 (SHC1), called p66Shc, and activates the extracellular signal-related kinase (ERK) pathway. Mice with adipocyte-specific OSMR deletion (OsmrFKO) are insulin resistant and exhibit adipose tissue inflammation, suggesting that intact adipocyte OSM-OSMR signaling is necessary for maintaining adipose tissue health. How OSM affects specific adipocyte functions is still unclear. Here, we examined the effects of OSM on adipocyte lipolysis. We treated 3T3-L1 adipocytes with OSM, insulin, and/or inhibitors of SHC1 and ERK and measured glycerol release. We also measured phosphorylation of p66Shc, ERK, and insulin receptor substrate-1 (IRS1) and the expression of lipolysis-associated genes in OSM-exposed 3T3-L1 adipocytes and primary adipocytes from control and OsmrFKO mice. We found that OSM induces adipocyte lipolysis via a p66Shc-ERK pathway and inhibits the suppression of lipolysis by insulin. Further, OSM induces phosphorylation of inhibitory IRS1 residues. We conclude that OSM is a stimulator of lipolysis and inhibits adipocyte insulin response. Future studies will determine how these roles of OSM affect adipose tissue function in health and disease.


Sujet(s)
Insuline , Lipolyse , Oncostatine M , Cellules 3T3-L1/métabolisme , Adipocytes/métabolisme , Animaux , Extracellular Signal-Regulated MAP Kinases/métabolisme , Insuline/métabolisme , Insuline ordinaire humaine , Lipolyse/effets des médicaments et des substances chimiques , Souris , Oncostatine M/métabolisme , Protéine transformante-1 contenant un domaine d'homologie-2 de Src/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...