Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 13.378
Filtrer
1.
Eur J Pharmacol ; 977: 176742, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38880216

RÉSUMÉ

Polycystic ovary syndrome (PCOS), a common endocrine disorder affecting premenopausal women, is associated with various metabolic consequences such as insulin resistance, hyperlipidemia, obesity, and type 2 diabetes mellitus (T2DM). Insulin sensitizers, such as metformin and pioglitazone, though effective, often leads to significant gastrointestinal adverse effects or weight gain, limiting its suitability for women with PCOS. There is an urgent need for safe, effective and affordable agents. Dapagliflozin, a sodium-glucose co-transporter 2 (SGLT2) inhibitor, enhances glucose elimination through urine, thereby reducing body weight and improving glucose and lipid metabolism. Nevertheless, it is not currently recommended as a therapeutic option for PCOS in clinical guidelines. In this study, we systematically examined the impact of dapagliflozin on an obese PCOS mouse model, focusing on alterations in glucose metabolism, adipose tissue morphology, and plasma lipid profile. Obese PCOS was induced in mice by continuous dihydrotestosterone (DHEA) injections over 21 days and high-fat diet (HFD) feeding. PCOS mice were then orally gavaged with dapagliflozin (1 mg/kg), metformin (50 mg/kg), or vehicle daily for 8 weeks, respectively. Our results demonstrated that dapagliflozin significantly prevented body weight gain and reduced fat mass in obese PCOS mice. Meanwhile, dapagliflozin treatment improved glucose tolerance and increased insulin sensitivity compared to the control PCOS mice. Furthermore, dapagliflozin significantly improved adipocyte accumulation and morphology in white adipose tissue, resulting in a normalized plasma lipid profile in PCOS mice. In conclusion, our results suggest that dapagliflozin is an effective agent in managing glucose and lipid metabolism disorders in obese PCOS mice.


Sujet(s)
Composés benzhydryliques , Glucosides , Insulinorésistance , Obésité , Syndrome des ovaires polykystiques , Animaux , Syndrome des ovaires polykystiques/traitement médicamenteux , Syndrome des ovaires polykystiques/métabolisme , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Composés benzhydryliques/pharmacologie , Composés benzhydryliques/usage thérapeutique , Femelle , Souris , Obésité/traitement médicamenteux , Obésité/métabolisme , Obésité/complications , Alimentation riche en graisse/effets indésirables , Modèles animaux de maladie humaine , Souris obèse , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/métabolisme , Metformine/pharmacologie , Metformine/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Métabolisme lipidique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Glycémie/effets des médicaments et des substances chimiques , Glycémie/métabolisme
2.
Eur J Pharmacol ; 977: 176745, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38880220

RÉSUMÉ

High fat diet (HFD) consumption can cause dysregulation of glucose and lipid metabolism, coupled with increased ectopic lipid deposition in renal tissue leading to steatosis and dysfunction. Sitagliptin is a dipeptidyl peptidase-4 (DPP-4) inhibitor clinically used for type II diabetes therapy; however its effect on renal steatosis in obese state is still uncertain. Herein, obesity was induced by feeding male Wistar rats HFD for 18 weeks, thereafter received either drug vehicle, or sitagliptin (10 mg/kg, PO) along with HFD for further 6 weeks and compared with age-matched rats receiving normal chow diet (NCD). After 24 weeks, serum and kidneys were collected for histological and biochemical assessments. Compared to NCD-fed group, HFD-fed rats displayed marked weight gain, increased fat mass, insulin resistance, dyslipidemia, impaired kidney functions and renal histological alterations. Sitagliptin effectively ameliorated obesity and related metabolic perturbations and improved kidney architecture and function. There were increased levels of triglycerides and cluster of differentiation 36 (CD36) in kidneys of obese rats, that were lowered by sitagliptin therapy. Sitagliptin significantly repressed the expression of lipogenesis genes, while up-regulated genes involved in mitochondrial biogenesis and fatty acid oxidation in kidneys of HFD-fed rats. Sitagliptin was found to induce down-regulation of endoplasmic reticulum (ER) stress and apoptotic markers in kidneys of obese rats. These findings together may emphasize a novel concept that sitagliptin can be an effective therapeutic approach for halting obesity-related renal steatosis and CKD.


Sujet(s)
Antigènes CD36 , Alimentation riche en graisse , Stress du réticulum endoplasmique , Rein , Obésité , Rat Wistar , Transduction du signal , Phosphate de sitagliptine , Animaux , Phosphate de sitagliptine/pharmacologie , Phosphate de sitagliptine/usage thérapeutique , Alimentation riche en graisse/effets indésirables , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Mâle , Obésité/traitement médicamenteux , Obésité/métabolisme , Obésité/complications , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Inhibiteurs de la dipeptidyl-peptidase IV/pharmacologie , Inhibiteurs de la dipeptidyl-peptidase IV/usage thérapeutique , Maladies du rein/traitement médicamenteux , Maladies du rein/métabolisme , Maladies du rein/étiologie , Maladies du rein/anatomopathologie , Maladies du rein/prévention et contrôle
3.
Eur J Pharmacol ; 977: 176705, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38830457

RÉSUMÉ

Obesity is a major health issue that contributes significantly to increased mortality and morbidity worldwide. Obesity is caused by uncontrolled adipogenesis and lipogenesis, leading to several metabolism-associated problems. Pancreatic lipase, an enzyme that breaks down dietary lipids, is a prominent target for obesity. Orlistat, a known inhibitor of pancreatic lipase, is commonly employed for the management of obesity. However, its side effects, such as diarrhoea, nausea and bladder pain, urge to look out for safer alternatives. Morin is a pentahydroxyflavone, exerts a broad spectrum of pharmacological effects including antioxidant, anti-inflammatory, lipid lowering, anti-diabetic, anti-fibrotic, anti-cancer, etc. This study investigated the effect of morin on pancreatic lipase activity, in vitro and in vivo adipogenesis. Molecular docking and simulation studies showed morin to have a higher binding affinity towards pancreatic lipase compared with orlistat, which also inhibited its activity in vitro. Morin also reduced lipid droplet accretion and downregulated the expression of adipogenic and lipogenic genes. The acute oral toxicity of morin was determined in C57BL/6 mice, where morin did not show toxicity up to 2000 mg/kg body weight dose. Oral administration of morin to high fat diet fed mice reduced body weight, glucose and insulin levels. Also, the histopathological examination revealed reduction in adipocyte size and decreased mRNA expression of adipogenesis markers in white adipose tissue of morin administered group compared to high fat diet group. Overall, the results suggested morin inhibited pancreatic lipase activity, adipogenesis and further studies are warranted to explore its therapeutic potential for obesity.


Sujet(s)
Adipogenèse , Flavonoïdes , Triacylglycerol lipase , Souris de lignée C57BL , Simulation de docking moléculaire , Animaux , Adipogenèse/effets des médicaments et des substances chimiques , Flavonoïdes/pharmacologie , Souris , Triacylglycerol lipase/antagonistes et inhibiteurs , Triacylglycerol lipase/métabolisme , Mâle , Cellules 3T3-L1 , Alimentation riche en graisse/effets indésirables , Pancréas/effets des médicaments et des substances chimiques , Pancréas/anatomopathologie , Agents antiobésité/pharmacologie , Obésité/traitement médicamenteux , Obésité/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Humains , Orlistat/pharmacologie , Flavones
4.
Nutrients ; 16(12)2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38931172

RÉSUMÉ

Obesity and type 2 diabetes are prevalent metabolic diseases that have significant links to several chronic diseases, including cancer, diabetes, hypertension, and cardiovascular disease. Muscadine grape extracts have shown the potential to reduce adiposity and improve insulin sensitivity and glucose control. Thus, this study was designed to determine the potential of muscadine grape berries extract (Pineapple and Southern Home) for its antiobesity properties in 3T3-L1 cells as a model for obesity research. The current study's data indicated the total phenolic content (TPC) and 2,2-diphenyl-1-picrylhydraziyl (DPPH) activity were higher in cultivar (CV) Southern Home, meanwhile, elevated the total flavonoid content (TFC) in Pineapple. Both extracts were safe across the tested range (0-5 mg/mL). A noticeable reduction in lipid accumulation was also found in extract-treated cells. In preadipocytes and adipocytes, the tested extracts showed significant alterations in various genes involved in glucose homeostasis and obesity. The most remarkable findings of the current study are the upregulation of two genes, Cntfr (+712.715-fold) and Hrh1 (+270.11-fold) in CV Pineapple extract-treated adipocytes 3T3-L1 and the high fold increase in Ramp3 induced by both Pineapple and Southern Home in pre-adipose cells. Furthermore, the tested extracts showed a potential to alter the mRNA of various genes, including Zfp91, B2m, Nr3c1, Insr, Atrn, Il6ra, Hsp90ab1, Sort1, and Npy1r. In conclusion, the data generated from the current study suggested that the two extracts under investigation are considered potential candidates for controlling insulin levels and managing obesity.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Agents antiobésité , Obésité , Extraits de plantes , Vitis , Animaux , Souris , Extraits de plantes/pharmacologie , Agents antiobésité/pharmacologie , Obésité/métabolisme , Obésité/traitement médicamenteux , Obésité/génétique , Vitis/composition chimique , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Fruit/composition chimique
5.
Nutrients ; 16(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38931268

RÉSUMÉ

Obesity is acknowledged as a significant risk factor for cardiovascular disease, often accompanied by increased inflammation and diabetes. Bioactive peptides derived from marine animal proteins show promise as safe and effective anti-obesity agents by regulating adipocyte differentiation through the AMPK signaling pathway. Therefore, this study aims to investigate the anti-obesity and anti-diabetic effects of bioactive compounds derived from a Meretrix lusoria Protamex enzymatic hydrolysate (MLP) fraction (≤1 kDa) through a 6-week treatment (150 mg/kg or 300 mg/kg, administered once daily) in leptin receptor-deficient db/db mice. The MLP treatment significantly decreased the body weight, serum total cholesterol, triglycerides, and LDL-cholesterol levels while also exhibiting a beneficial effect on hepatic and serum marker parameters in db/db mice. A histological analysis revealed a reduction in hepatic steatosis and epididymal fat following MLP treatment. Furthermore, poor glucose tolerance was improved, and hepatic antioxidant enzyme activities were elevated in MLP-treated mice compared to db/db control mice. Western blot analysis showed an increased expression of the AMPK protein after MLP treatment. In addition, the expression of lipogenic genes decreased in db/db mice. These findings indicate that bioactive peptides, which are known to regulate blood glucose levels, lipid metabolism, and adipogenesis, could be beneficial functional food additives and pharmaceuticals.


Sujet(s)
Agents antiobésité , Obésité , Peptides , Animaux , Obésité/traitement médicamenteux , Souris , Mâle , Peptides/pharmacologie , Agents antiobésité/pharmacologie , Hydrolysats de protéines/pharmacologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Glycémie/métabolisme , Hypoglycémiants/pharmacologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme , Souris de lignée C57BL , Récepteurs à la leptine/métabolisme , Récepteurs à la leptine/génétique , Adipogenèse/effets des médicaments et des substances chimiques , Poids/effets des médicaments et des substances chimiques
6.
Nutrients ; 16(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38931313

RÉSUMÉ

Obesity is increasingly prevalent worldwide and is linked to metabolic diseases, such as insulin resistance (IR) and type 2 diabetes mellitus (T2DM), due to excessive free fatty acids (FFAs). Although lifestyle changes are effective, they often prove to be insufficient as initial treatments for obesity. Additionally, while surgical and pharmacological interventions are available, they are not entirely safe or effective. Recently, interest has grown in utilizing food waste and plant-derived phenolic compounds for their health benefits, presenting a promising avenue for managing obesity and its related disorders. Indeed, many studies have examined the potential inhibitory effects of the natural extract on adipocyte differentiation and lipid accumulation. This study focused on the evaluation of the effects of standardized extracts obtained from red oranges and olive leaf waste on 3T3-L1 murine pre-adipocyte and adipocyte functionality. Red orange extract (ROE) and olive leaf extract (OLE), alone and in combination, were tested to assess their anti-obesity and anti-inflammatory effects, as well as their potential therapeutic benefits. Three in vitro models were established to investigate the effects of the extracts on (I) adipocyte differentiation; (II) mature and hypertrophic adipocytes challenged with palmitic acid (PA) and erastin (ER), respectively; and (III) erastin-induced cytotoxicity on pre-adipocytes.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Olea , Extraits de plantes , Feuilles de plante , Animaux , Olea/composition chimique , Adipocytes/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Souris , Feuilles de plante/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Agents antiobésité/pharmacologie , Adipogenèse/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux
7.
J Food Drug Anal ; 32(2): 213-226, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38934690

RÉSUMÉ

Citrus peels contain abundant polyphenols, particularly flavonoids, and have been shown to exert lipid accumulation decreasing ability. In this study, Citrus depressa peel applied to oven drying and extracted with ethanol extract as CDEE to analyze its flavonoids compositions and investigated its effects on a high-fat diet (HFD)-induced obese mice model. CDEE contained several flavonoids such as hesperidin, sinesentin, nobiletin, tangeretin, 5-demethylnobiletin, and 5-demethyltangeretin. The mice fed an HFD, and administration of 2% CDEE to could decrease weight gain, abdominal fat weight, inguinal fat weight, and the adipocyte size, and CDEE also reduced serum total cholesterol (TCHO), triacylglycerol (TG) compared with mice fed only on HFD. CDEE hindered lipid accumulation through a decreased fatty acid synthase (FAS) protein expression via upregulation of the protein expression of AMP-activated protein kinase α (AMPKα). Moreover, CDEE modulated gut microbiota that altered by HFD through an increased abundance of Lactobacillus reuteri compared with the HFD group. The results demonstrated that CDEE helps decrease lipid accumulation through the AMPK pathway, which also indicates a prebiotic-like effect on gut microbiota.


Sujet(s)
Citrus , Alimentation riche en graisse , Microbiome gastro-intestinal , Métabolisme lipidique , Souris de lignée C57BL , Souris obèse , Obésité , Extraits de plantes , Prébiotiques , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie , Extraits de plantes/composition chimique , Citrus/composition chimique , Mâle , Obésité/métabolisme , Obésité/traitement médicamenteux , Métabolisme lipidique/effets des médicaments et des substances chimiques , Prébiotiques/administration et posologie , Prébiotiques/analyse , Alimentation riche en graisse/effets indésirables , Humains , Triglycéride/métabolisme , Triglycéride/sang , Bactéries/classification , Bactéries/isolement et purification , Bactéries/génétique , Bactéries/métabolisme , Bactéries/effets des médicaments et des substances chimiques
8.
J Food Drug Anal ; 32(2): 227-238, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38934691

RÉSUMÉ

We aimed to investigate the therapeutic potential of ibuprofen against type 2 diabetes (T2D) using obese Zucker diabetic fatty (ZDF) rats as type 2 diabetes model. ZDF rats were hyperglycemic, dyslipidemic and expressed proinflammatory markers in contrast to lean controls, thus reflecting the relationship between obesity and chronic inflammation promoting T2D. Chronic treatment with ibuprofen (2-(4-Isobutylphenyl)propanoic acid) was used to study the impact on pathological T2D conditions as compared to metformin (1,1-dimethylbiguanide) treated ZDF as well as lean controls. Ibuprofen decreased A1c but induced a high insulin release with improved glucose tolerance only after early time points (i.g., 15 and 30 min) resulting in a non-significant decline of AUC values and translating into a high HOMA-IR. In addition, ibuprofen significantly lowered cholesterol, free fatty acids and HDL-C. Some of these effects by ibuprofen might be based on its anti-inflammatory effects through inhibition of cytokine/chemokine signaling (i.g., COX-2, ICAM-1 and TNF-α) as measured in whole blood and epididymal adipose tissue by TaqMan and/or upregulation of anti-inflammatory cytokines (i.g., IL-4 and IL-13) by ELISA analysis in blood. In conclusion, our ZDF animal study showed positive effects of ibuprofen against diabetic complications such as inflammation and dyslipidemia but also demonstrated the risk of causing insulin resistance.


Sujet(s)
Diabète de type 2 , Ibuprofène , Rat Zucker , Animaux , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Ibuprofène/pharmacologie , Ibuprofène/administration et posologie , Rats , Mâle , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Humains , Modèles animaux de maladie humaine , Insuline/métabolisme , Obésité/traitement médicamenteux , Obésité/métabolisme , Cytokines/métabolisme , Insulinorésistance
9.
In Vivo ; 38(4): 1557-1570, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936927

RÉSUMÉ

BACKGROUND/AIM: This study examined the effects of tocotrienols (TT) in conjunction with statin on glucose homeostasis, bone microstructure, gut microbiome, and systemic and liver inflammatory markers in obese C57BL/6J mice. MATERIALS AND METHODS: Forty male C57BL/6J mice were fed a high-fat diet (HFD) and assigned into four groups in a 2 (no statin vs. 120 mg statin/kg diet)×2 (no TT vs. 400 mg TT/kg diet) factorial design for 14 weeks. RESULTS: Statin and TT improved glucose tolerance only when each was given alone, and only statin supplementation decreased insulin resistance. Consistently, only statin supplementation decreased serum insulin levels and HOMA-IR. Pancreatic insulin was also increased with statin treatment. Statin and TT, alone or in combination, reduced the levels of serum IL-6, but only TT attenuated the increased serum leptin levels induced by a HFD. Statin supplementation increased bone area/total area and connectivity density at LV-4, while TT supplementation increased bone area/total area and trabecular number, but decreased trabecular separation at the distal femur. Statin supplementation, but not TT, reduced hepatic inflammatory cytokine gene expression. Neither TT supplementation nor statin supplementation statistically altered microbiome species evenness or richness. However, they altered the relative abundance of certain microbiome species. Most notably, both TT and statin supplementation increased the relative abundance of Lachnospiraceae UCG-006. CONCLUSION: TT and statin collectively benefit bone microstructure, glucose homeostasis, and microbial ecology in obese mice. Such changes may be, in part, associated with suppression of inflammation in the host.


Sujet(s)
Os et tissu osseux , Alimentation riche en graisse , Compléments alimentaires , Microbiome gastro-intestinal , Homéostasie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Obésité , Tocotriénols , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Tocotriénols/pharmacologie , Tocotriénols/administration et posologie , Souris , Homéostasie/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Obésité/métabolisme , Mâle , Os et tissu osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/métabolisme , Os et tissu osseux/anatomopathologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Alimentation riche en graisse/effets indésirables , Bixaceae/composition chimique , Souris obèse , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie , Glucose/métabolisme , Souris de lignée C57BL , Insulinorésistance , Glycémie , Modèles animaux de maladie humaine , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Marqueurs biologiques , Caroténoïdes
10.
Biomolecules ; 14(6)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38927022

RÉSUMÉ

Recent studies increasingly suggest that targeting brown/beige adipose tissues to enhance energy expenditure offers a novel therapeutic approach for treating metabolic diseases. Brown/beige adipocytes exhibit elevated expression of uncoupling protein 1 (UCP1), which is a thermogenic protein that efficiently converts energy into heat, particularly in response to cold stimulation. Polyphenols possess potential anti-obesity properties, but their pharmacological effects are limited by their bioavailability and distribution within tissue. This study discovered 18a, a polyphenol compound with a favorable distribution within adipose tissues, which transcriptionally activates UCP1, thereby promoting thermogenesis and enhancing mitochondrial respiration in brown adipocytes. Furthermore, in vivo studies demonstrated that 18a prevents high-fat-diet-induced weight gain and improves insulin sensitivity. Our research provides strong mechanistic evidence that UCP1 is a complex mediator of 18a-induced thermogenesis, which is a critical process in obesity mitigation. Brown adipose thermogenesis is triggered by 18a via the AMPK-PGC-1α pathway. As a result, our research highlights a thermogenic controlled polyphenol compound 18a and clarifies its underlying mechanisms, thus offering a potential strategy for the thermogenic targeting of adipose tissue to reduce the incidence of obesity and its related metabolic problems.


Sujet(s)
Alimentation riche en graisse , Obésité , Polyphénols , Thermogenèse , Protéine-1 de découplage , Protéine-1 de découplage/métabolisme , Protéine-1 de découplage/génétique , Thermogenèse/effets des médicaments et des substances chimiques , Animaux , Obésité/métabolisme , Obésité/traitement médicamenteux , Polyphénols/pharmacologie , Souris , Alimentation riche en graisse/effets indésirables , Tissu adipeux brun/métabolisme , Tissu adipeux brun/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Humains , Métabolisme énergétique/effets des médicaments et des substances chimiques
11.
Ren Fail ; 46(1): 2351473, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38915241

RÉSUMÉ

OBJECTIVE: Liraglutide, a glucagon-like peptide-1 receptor agonist, has been shown to regulate blood sugar and control body weight, but its ability to treat obesity-related nephropathy has been poorly studied. Therefore, this study was designed to observe the characteristics and potential mechanism of liraglutide against obesity-related kidney disease. METHODS: Thirty-six C57BL/6J male mice were randomly divided into six groups (n = 6 per group). Obesity-related nephropathy was induced in mice by continuous feeding of high-fat diet (HFD) for 12 weeks. After 12 weeks, liraglutide (0.6 mg/kg) and AMP-activated protein kinase (AMPK) agonists bortezomib (200 µg/kg) were injected for 12 weeks, respectively. Enzyme-linked immunosorbent assay was employed to detect the levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, blood urea nitrogen, creatinine in serum, as well as urinary protein in urine. Besides, hematoxylin-eosin staining and periodic acid-Schiff staining were used to observe the pathological changes of kidney tissue; immunohistochemistry, western blot, and real-time quantitative PCR to assess the calmodulin-dependent protein kinase kinase beta (CaMKKß)/AMPK signaling pathway activation. RESULTS: Liraglutide significantly reduced serum lipid loading, improved kidney function, and relieved kidney histopathological damage and glycogen deposition in the mouse model of obesity-related kidney disease induced by HFD. In addition, liraglutide also significantly inhibited the CaMKKß/AMPK signaling pathway in kidney tissue of HFD-induced mice. However, bortezomib partially reversed the therapeutic effect of liraglutide on HDF-induced nephropathy in mice. CONCLUSIONS: Liraglutide has a therapeutic effect on obesity-related kidney disease, and such an effect may be achieved by inhibiting the CaMKKß/AMPK signaling pathway in kidney tissue.


Sujet(s)
AMP-Activated Protein Kinases , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Alimentation riche en graisse , Liraglutide , Souris de lignée C57BL , Obésité , Transduction du signal , Animaux , Liraglutide/pharmacologie , Liraglutide/usage thérapeutique , Mâle , Alimentation riche en graisse/effets indésirables , Souris , AMP-Activated Protein Kinases/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Kinase/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Obésité/complications , Obésité/traitement médicamenteux , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Modèles animaux de maladie humaine , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique
12.
JAMA Netw Open ; 7(6): e2416775, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38916894

RÉSUMÉ

Importance: A major concern with weight loss is concomitant bone loss. Exercise and glucagon-like peptide-1 receptor agonists (GLP-1RAs) represent weight loss strategies that may protect bone mass despite weight loss. Objective: To investigate bone health at clinically relevant sites (hip, spine, and forearm) after diet-induced weight loss followed by a 1-year intervention with exercise, liraglutide, or both combined. Design, Setting, and Participants: This study was a predefined secondary analysis of a randomized clinical trial conducted between August 2016 and November 2019 at the University of Copenhagen and Hvidovre Hospital in Denmark. Eligible participants included adults aged 18 to 65 years with obesity (body mass index of 32-43) and without diabetes. Data analysis was conducted from March to April 2023, with additional analysis in February 2024 during revision. Interventions: After an 8-week low-calorie diet (800 kcal/day), participants were randomized to 1 of 4 groups for 52 weeks: a moderate- to vigorous-intensity exercise program (exercise alone), 3.0 mg daily of the GLP-1 RA liraglutide (liraglutide alone), the combination, or placebo. Main Outcomes and Measures: The primary outcome was change in site-specific bone mineral density (BMD) at the hip, lumbar spine, and distal forearm from before the low-calorie diet to the end of treatment, measured by dual-energy x-ray absorptiometry in the intention-to-treat population. Results: In total, 195 participants (mean [SD] age, 42.84 [11.87] years; 124 female [64%] and 71 male [36%]; mean [SD] BMI, 37.00 [2.92]) were randomized, with 48 participants in the exercise group, 49 participants in the liraglutide group, 49 participants in the combination group, and 49 participants in the placebo group. The total estimated mean change in weight losses during the study was 7.03 kg (95% CI, 4.25-9.80 kg) in the placebo group, 11.19 kg (95% CI, 8.40-13.99 kg) in the exercise group, 13.74 kg (95% CI, 11.04-16.44 kg) in the liraglutide group, and 16.88 kg (95% CI, 14.23-19.54 kg) in the combination group. In the combination group, BMD was unchanged compared with the placebo group at the hip (mean change, -0.006 g/cm2; 95% CI, -0.017 to 0.004 g/cm2; P = .24) and lumbar spine (-0.010 g/cm2; 95% CI, -0.025 to 0.005 g/cm2; P = .20). Compared with the exercise group, BMD decreased for the liraglutide group at the hip (mean change, -0.013 g/cm2; 95% CI, -0.024 to -0.001 g/cm2; P = .03) and spine (mean change, -0.016 g/cm2; 95% CI, -0.032 to -0.001 g/cm2; P = .04). Conclusions and Relevance: In this randomized clinical trial, the combination of exercise and GLP-1RA (liraglutide) was the most effective weight loss strategy while preserving bone health. Liraglutide treatment alone reduced BMD at clinically relevant sites more than exercise alone despite similar weight loss. Trial Registration: EudraCT: 2015-005585-32.


Sujet(s)
Densité osseuse , Exercice physique , Récepteur du peptide-1 similaire au glucagon , Liraglutide , Humains , Femelle , Mâle , Adulte d'âge moyen , Liraglutide/usage thérapeutique , Récepteur du peptide-1 similaire au glucagon/agonistes , Densité osseuse/effets des médicaments et des substances chimiques , Adulte , Obésité/traitement médicamenteux , Obésité/thérapie , Perte de poids/effets des médicaments et des substances chimiques , Hypoglycémiants/usage thérapeutique , Sujet âgé , Association thérapeutique , Danemark
13.
Physiol Rep ; 12(12): e16085, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38924673

RÉSUMÉ

Methylphenidate (MPH) has been previously shown to increase resting energy expenditure (REE) in individuals of normal weight; however, the effects on individuals living with obesity are currently unknown. Ten individuals living with obesity were randomly assigned to undergo 60 days of MPH administration with a daily dose of 0.5 mg/kg body weight or a placebo control. REE was measured before and after the 60-day intervention. There was a trend toward significance for group × time interaction on REE (p = 0.082) with a large effect size (η2 = 0.331), with MPH administration increasing REE compared to a decrease in placebo control. Preliminary findings from this pilot study show that MPH has the potential to counter the adaptive thermogenic process commonly seen in weight loss. This is a unique finding among pharmacotherapies, as no approved obesity drugs measurably impact REE.


Sujet(s)
Métabolisme énergétique , Méthylphénidate , Obésité , Humains , Méthylphénidate/usage thérapeutique , Méthylphénidate/pharmacologie , Mâle , Femelle , Obésité/métabolisme , Obésité/traitement médicamenteux , Projets pilotes , Métabolisme énergétique/effets des médicaments et des substances chimiques , Adulte , Méthode en double aveugle , Adulte d'âge moyen , Stimulants du système nerveux central/usage thérapeutique , Stimulants du système nerveux central/pharmacologie
14.
Hastings Cent Rep ; 54(3): 6-10, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38842904

RÉSUMÉ

New anti-obesity medications (AOMs) have received widespread acclaim in medical journals and the media, but they also raise critical ethical, public health, and public policy concerns that have largely been ignored. AOMs are very costly, need to be taken by a patient in perpetuity (since significant rebound weight gain otherwise occurs), and threaten to shift resources and focus away from other crucial efforts at obesity treatment and prevention. Many people may feel less motivated to exercise or reduce their caloric consumption, if they assume that obesity is now medically treatable. Policy-makers may similarly come to feel that the solution to the obesity pandemic is simply to prescribe medications and that prevention efforts are far less necessary. These drugs raise concerns about justice (since AOMs will disproportionately benefit the wealthy), medicalization, and marketing. Policy-makers, clinicians, and others need to engage in multipronged educational and policy efforts to address these challenges.


Sujet(s)
Agents antiobésité , Politique de santé , Obésité , Santé publique , Humains , Obésité/traitement médicamenteux , Santé publique/éthique , États-Unis
16.
Sci Rep ; 14(1): 12685, 2024 06 03.
Article de Anglais | MEDLINE | ID: mdl-38830962

RÉSUMÉ

White kidney bean (Phaseolus vulgaris L.) extracts can aid weight management by reducing calorie intake from complex carbohydrates through alpha-amylase inhibition. We examined the impact of a proprietary aqueous extract from whole dried white kidney beans standardized by its alpha-amylase inhibitor activity (Phase 2 white kidney bean extract (WKBE)) on weight management in subjects with overweight and moderate obesity. In a randomized, double-blind, placebo-controlled fashion, 81 participants completed the study and ingested either a high dose of Phase 2 (1000 mg, WKBE HIGH), a low dose (700 mg, WKBE LOW), or a matching placebo (microcrystalline cellulose, PLA) three times a day, 30 min before meals, for 12 weeks during a calorie restricted diet. In a dose-dependent manner, Phase 2 significantly reduced body weight, fat mass, BMI, waist, hip and in the WKBE HIGH group thigh circumference. Phase 2 is an effective and safe supplement aiding weight and fat loss. ClinicalTrials.gov identifier NCT02930668.


Sujet(s)
Phaseolus , Extraits de plantes , Humains , Mâle , Femelle , Méthode en double aveugle , Phaseolus/composition chimique , Adulte d'âge moyen , Adulte , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Perte de poids/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , alpha-Amylases/antagonistes et inhibiteurs , alpha-Amylases/métabolisme , Surpoids/traitement médicamenteux , Lectines végétales
17.
Ann Endocrinol (Paris) ; 85(3): 179-183, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38871515

RÉSUMÉ

During the past years, several drugs have been developed for the treatment of obesity. Some are already used in clinical practice: orlistat, GLP-1 receptor agonists (RA), GLP-1/GIP biagonists and the melanocortin 4 receptor (MC4R) agonist, setmelanotide. Some should be available in the future: GLP-1/glucagon biagonists, GLP-1/GIP/glucagon triagonists. These drugs act mainly by reducing food intake or fat absorption. However, many of them show specific effects on the adipose tissue. All these drugs show significant reduction of fat mass and, more particularly of visceral fat. If most of the drugs, except orlistat, have been shown to increase energy expenditure in rodents with enhanced thermogenesis, this has not yet been clearly demonstrated in humans. However, biagonists or triagonist stimulating glucagon seem to a have a more potent effect to increase thermogenesis in the adipose tissue and, thus, energy expenditure. Most of these drugs have been shown to increase the production of adiponectin and to reduce the production of pro-inflammatory cytokines by the adipose tissue. GLP-1RAs reduce the size of adipocytes and promote their differentiation. GLP-1RAS and GLP-1/GIP biagonists reduce, in the adipose tissue, the expression of several genes involved in lipogenesis. Further studies are still needed to clarify the precise roles, on the adipose tissue, of these drugs dedicated for the treatment of obesity.


Sujet(s)
Tissu adipeux , Agents antiobésité , Métabolisme énergétique , Obésité , Humains , Obésité/traitement médicamenteux , Obésité/métabolisme , Agents antiobésité/usage thérapeutique , Agents antiobésité/pharmacologie , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/métabolisme , Animaux , Métabolisme énergétique/effets des médicaments et des substances chimiques , Récepteur du peptide-1 similaire au glucagon/agonistes , Thermogenèse/effets des médicaments et des substances chimiques , Thermogenèse/physiologie , Glucagon-like peptide 1/agonistes , Orlistat/usage thérapeutique , Orlistat/pharmacologie
19.
Molecules ; 29(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38893431

RÉSUMÉ

BACKGROUND: With the changes in lifestyle and diet structure, the incidence of obesity has increased year by year, and obesity is one of the inducements of many chronic metabolic diseases. Epigallocatechin gallate (EGCG), which is the most abundant component of tea polyphenols, has been used for many years to improve obesity and its complications. Though it has been reported that EGCG can improve obesity through many molecular mechanisms, EGCG may have many mechanisms yet to be explored. In this study, we explored other possible mechanisms through molecular docking and in vitro experiments. METHODS: AutoDock Vina was selected for conducting the molecular docking analysis to elucidate the interaction between EGCG and Notch1, while molecular dynamics simulations were employed to validate this interaction. Then, the new regulation mechanism of EGCG on obesity was verified with in vitro experiments, including a Western blot experiment, immunofluorescence experiment, oil red O staining, and other experiments in 3T3-L1 adipocytes. RESULTS: The molecular docking results showed that EGCG could bind to Notch1 protein through hydrogen bonding. In vitro cell experiments demonstrated that EGCG can significantly reduce the sizes of lipid droplets of 3T3-L1 adipocytes and promote UCP-1 expression by inhibiting the expression of Notch1 in 3T3-L1 adipocytes, thus promoting mitochondrial biogenesis. CONCLUSIONS: In this study, molecular docking and in vitro cell experiments were used to explore the possible mechanism of EGCG to improve obesity by inhibiting Notch1.


Sujet(s)
Adipogenèse , Catéchine , Simulation de docking moléculaire , Récepteur Notch1 , Animaux , Souris , Cellules 3T3-L1 , Adipogenèse/effets des médicaments et des substances chimiques , Catéchine/analogues et dérivés , Catéchine/pharmacologie , Catéchine/composition chimique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Simulation de dynamique moléculaire , Obésité/traitement médicamenteux , Obésité/métabolisme , Récepteur Notch1/métabolisme , Protéine-1 de découplage/métabolisme
20.
Expert Rev Cardiovasc Ther ; 22(6): 217-230, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38864827

RÉSUMÉ

INTRODUCTION: Heart failure with preserved ejection fraction (HFpEF) is a highly heterogeneous syndrome, making it challenging to improve prognosis with pharmacotherapy. Obesity is one of the leading phenotypes of HFpEF, and its prevalence continues to grow worldwide. Consequently, obesity-targeted interventions have attracted attention as a novel treatment strategy for HFpEF. AREAS COVERED: The authors review the association between the pathogenesis of obesity and HFpEF and the potential for obesity-targeted pharmacotherapeutic strategies in HFpEF, together with the latest evidence. The literature search was conducted in PubMed up to April 2024. EXPERT OPINION: The STEP HFpEF (Semaglutide Treatment Effect in People with obesity and HFpEF) and SELECT (Semaglutide Effects on Cardiovascular Outcomes in People with Overweight or Obesity) trials recently demonstrated that the glucagon-like peptide 1 analogue, semaglutide, improves various aspects of clinical outcomes in obese HFpEF patients and significantly reduces cardiovascular and heart failure events in non-diabetic obese patients, along with a substantial weight loss. Future clinical trials with other incretin mimetics with more potent weight loss and sub-analyses of the SELECT trial may further emphasize the importance of the obesity phenotype-based approach in the treatment of HFpEF.


Sujet(s)
Défaillance cardiaque , Obésité , Débit systolique , Perte de poids , Humains , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Obésité/complications , Obésité/traitement médicamenteux , Débit systolique/effets des médicaments et des substances chimiques , Perte de poids/effets des médicaments et des substances chimiques , Agents antiobésité/usage thérapeutique , Agents antiobésité/pharmacologie , Pronostic , Animaux , Peptides glucagon-like
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...