Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37.035
Filtrer
1.
Nutrients ; 16(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38931174

RÉSUMÉ

Zinc deficiency has been associated with the worsening of diabetes while zinc supplementation has been proposed to ameliorate diabetes. This study examined the effects of marginal zinc deficiency (MZD) and zinc supplementation (ZS) on obesity, glycemic control, pancreatic islets, hepatic steatosis and renal function of Zucker diabetic fatty (ZDF) rats. Male ZDF rats were fed an MZD, zinc control (ZC) or ZS diet (4, 30 and 300 mg Zn/kg diet, respectively), and lean Zucker rats were fed a ZC diet for 8 weeks. MZD and ZS did not alter body weight or whole-body composition in ZDF rats. MZD ZDF rats had reduced zinc concentrations in the femur and pancreas, a greater number of enlarged pancreatic islets and a diminished response to an oral glucose load based on a 1.8-fold greater incremental area-under-the-curve (AUC) for glucose compared to ZC ZDF. ZS ZDF rats had elevated serum, femur and pancreatic zinc concentrations, unchanged pancreatic parameters and a 50% reduction in the AUC for insulin compared to ZC ZDF rats, suggesting greater insulin sensitivity. Dietary zinc intake did not alter hepatic steatosis, creatinine clearance, or levels of proteins that contribute to insulin signaling, inflammation or zinc transport in epididymal fat. Potential adverse effects of ZS were suggested by reduced hepatic copper concentrations and elevated serum urea compared to ZC ZDF rats. In summary, ZS improved the pancreatic insulin response but not the glucose handling. In contrast, reduced zinc status in ZDF rats led to impaired glucose tolerance and a compensatory increase in the number and size of pancreatic islets which could lead to ß-cell exhaustion.


Sujet(s)
Compléments alimentaires , Insuline , Ilots pancréatiques , Rat Zucker , Zinc , Animaux , Zinc/déficit , Mâle , Insuline/sang , Ilots pancréatiques/métabolisme , Ilots pancréatiques/effets des médicaments et des substances chimiques , Rats , Glycémie/métabolisme , Obésité/métabolisme , Insulinorésistance , Pancréas/métabolisme , Pancréas/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Diabète expérimental/traitement médicamenteux
2.
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
3.
Nutrients ; 16(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38931176

RÉSUMÉ

The global rise in type 2 diabetes (T2D) and obesity necessitates innovative dietary interventions. This study investigates the effects of allulose, a rare sugar shown to reduce blood glucose, in a rat model of diet-induced obesity and T2D. Over 12 weeks, we hypothesized that allulose supplementation would improve body weight, insulin sensitivity, and glycemic control. Our results showed that allulose mitigated the adverse effects of high-fat, high-sugar diets, including reduced body weight gain and improved insulin resistance. The allulose group exhibited lower food consumption and increased levels of glucagon-like peptide-1 (GLP-1), enhancing glucose regulation and appetite control. Additionally, allulose prevented liver triglyceride accumulation and promoted mitochondrial uncoupling in adipose tissue. These findings suggest that allulose supplementation can improve metabolic health markers, making it a promising dietary component for managing obesity and T2D. Further research is needed to explore the long-term benefits and mechanisms of allulose in metabolic disease prevention and management. This study supports the potential of allulose as a safe and effective intervention for improving metabolic health in the context of dietary excess.


Sujet(s)
Glycémie , Diabète de type 2 , Alimentation riche en graisse , Fructose , Insulinorésistance , Obésité , Animaux , Fructose/administration et posologie , Mâle , Obésité/métabolisme , Diabète de type 2/prévention et contrôle , Diabète de type 2/métabolisme , Glycémie/métabolisme , Rats , Alimentation riche en graisse/effets indésirables , Foie/métabolisme , Glucagon-like peptide 1/métabolisme , Glucagon-like peptide 1/sang , Triglycéride/sang , Rat Sprague-Dawley , Tissu adipeux/métabolisme , Prise de poids , Modèles animaux de maladie humaine
4.
Nutrients ; 16(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38931177

RÉSUMÉ

CONTEXT/OBJECTIVE: In order to better understand which metabolic differences are related to insulin resistance in metabolic syndrome (MetSyn), we used hyperinsulinemic-euglycemic (HE) clamps in individuals with MetSyn and related peripheral insulin resistance to circulating biomarkers. DESIGN/METHODS: In this cross-sectional study, HE-clamps were performed in treatment-naive men (n = 97) with MetSyn. Subjects were defined as insulin-resistant based on the rate of disappearance (Rd). Machine learning models and conventional statistics were used to identify biomarkers of insulin resistance. Findings were replicated in a cohort with n = 282 obese men and women with (n = 156) and without (n = 126) MetSyn. In addition to this, the relation between biomarkers and adipose tissue was assessed by nuclear magnetic resonance imaging. RESULTS: Peripheral insulin resistance is marked by changes in proteins related to inflammatory processes such as IL-1 and TNF-receptor and superfamily members. These proteins can distinguish between insulin-resistant and insulin-sensitive individuals (AUC = 0.72 ± 0.10) with MetSyn. These proteins were also associated with IFG, liver fat (rho 0.36, p = 1.79 × 10-9) and visceral adipose tissue (rho = 0.35, p = 6.80 × 10-9). Interestingly, these proteins had the strongest association in the MetSyn subgroup compared to individuals without MetSyn. CONCLUSIONS: MetSyn associated with insulin resistance is characterized by protein changes related to body fat content, insulin signaling and pro-inflammatory processes. These findings provide novel targets for intervention studies and should be the focus of future in vitro and in vivo studies.


Sujet(s)
Marqueurs biologiques , Insulinorésistance , Syndrome métabolique X , Protéome , Humains , Syndrome métabolique X/métabolisme , Mâle , Femelle , Études transversales , Adulte d'âge moyen , Adulte , Marqueurs biologiques/sang , Technique du clamp glycémique , Obésité/métabolisme , Tissu adipeux/métabolisme , Insuline/sang , Insuline/métabolisme , Graisse intra-abdominale/métabolisme
5.
Nutrients ; 16(12)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38931191

RÉSUMÉ

Skeletal muscle is composed of bundles of muscle fibers with distinctive characteristics. Oxidative muscle fiber types contain higher mitochondrial content, relying primarily on oxidative phosphorylation for ATP generation. Notably, as a result of obesity, or following prolonged exposure to a high-fat diet, skeletal muscle undergoes a shift in fiber type toward a glycolytic type. Mitochondria are highly dynamic organelles, constantly undergoing mitochondrial biogenesis and dynamic processes. Our study aims to explore the impact of obesity on skeletal muscle mitochondrial biogenesis and dynamics and also ascertain whether the skeletal muscle fiber type shift occurs from the aberrant mitochondrial machinery. Furthermore, we investigated the impact of exercise in preserving the oxidative muscle fiber types despite obesity. Mice were subjected to a normal standard chow and water or high-fat diet with sugar water (HFS) with or without exercise training. After 12 weeks of treatment, the HFS diet resulted in a noteworthy reduction in the markers of mitochondrial content, which was recovered by exercise training. Furthermore, higher mitochondrial biogenesis markers were observed in the exercised group with a subsequent increase in the mitochondrial fission marker. In conclusion, these findings imply a beneficial impact of moderate-intensity exercise on the preservation of oxidative capacity in the muscle of obese mouse models.


Sujet(s)
Alimentation riche en graisse , Modèles animaux de maladie humaine , Mitochondries du muscle , Muscles squelettiques , Obésité , Biogenèse des organelles , Conditionnement physique d'animal , Animaux , Obésité/métabolisme , Alimentation riche en graisse/effets indésirables , Conditionnement physique d'animal/physiologie , Muscles squelettiques/métabolisme , Souris , Mâle , Mitochondries du muscle/métabolisme , Souris de lignée C57BL , Marqueurs biologiques/métabolisme , Dynamique mitochondriale , Fibres musculaires squelettiques/métabolisme
6.
Med Sci Monit ; 30: e944462, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38932442

RÉSUMÉ

BACKGROUND Preliminary data suggest an adipogenic role for growth arrest-specific 6 (Gas6), a pleiotropic molecule involved in inflammation, proliferation, and hemostasis through its Tyro3, Axl, and MerTK (TAM) receptors. This study compares Gas6 expression in plasma and visceral and subcutaneous adipose tissue in 42 adults with obesity (body mass index ≥40 kg/m²) and 32 normal-weight controls to elucidate its role in obesity and related metabolic alterations. MATERIAL AND METHODS Using a case-control design, we measured Gas6 levels in plasma via a validated sandwich enzyme-linked immunosorbent assay and in adipose tissues through quantitative polymerase chain reactio with specific probes. Medians and correlations were analyzed using Mann-Whitney and Spearman tests. A general linear model assessed the impact of covariates on the Gas6-anthropometric relationship, with statistical significance determined by P values. RESULTS Plasma Gas6 levels were significantly higher in the obese group than in controls (P=0.0006). While Gas6 mRNA expression did not significantly differ in subcutaneous adipose tissue between groups, it was notably higher in visceral than subcutaneous adipose tissue in controls (P<0.05). A significant correlation was found between plasma Gas6 levels and body mass index (P=0.001). CONCLUSIONS Gas6 plasma levels are elevated in morbid obesity, particularly in visceral adipose tissue, and are linked to altered glucose tolerance in female patients. These findings highlight the role of Gas6 in obesity-related metabolic complications and suggest avenues for further research and potential therapies.


Sujet(s)
Tissu adipeux , Indice de masse corporelle , Inflammation , Protéines et peptides de signalisation intercellulaire , Obésité morbide , Humains , Femelle , Mâle , Adulte , Protéines et peptides de signalisation intercellulaire/sang , Protéines et peptides de signalisation intercellulaire/métabolisme , Inflammation/sang , Inflammation/métabolisme , Études cas-témoins , Adulte d'âge moyen , Tissu adipeux/métabolisme , Obésité morbide/sang , Obésité morbide/métabolisme , Graisse intra-abdominale/métabolisme , Graisse sous-cutanée/métabolisme , Obésité/métabolisme , Obésité/sang
7.
Obesity (Silver Spring) ; 32(7): 1373-1388, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38932722

RÉSUMÉ

OBJECTIVE: Obesity is characterized by dysregulated homeostatic mechanisms resulting in positive energy balance; however, when this dysregulation occurs is unknown. We assessed the time course of alterations to behaviors promoting weight gain in male and female mice switched to an obesogenic high-fat diet (HFD). METHODS: Male and female C57BL/6J mice were housed in metabolic chambers and were switched from chow to a 60% or 45% HFD for 4 and 3 weeks, respectively. Food intake, meal patterns, energy expenditure (EE), and body weight were continuously measured. A separate cohort of male mice was switched from chow to a 60% HFD and was given access to locked or unlocked running wheels. RESULTS: Switching mice to obesogenic diets promotes transient bouts of hyperphagia during the first 2 weeks followed by persistent caloric hyperphagia. EE increases but not sufficiently enough to offset increased caloric intake, resulting in a sustained net positive energy balance. Hyperphagia is associated with consumption of calorically larger meals (impaired satiation) more frequently (impaired satiety), particularly during the light cycle. Running wheel exercise delays weight gain in male mice fed a 60% HFD by enhancing satiation and increasing EE. However, exercise effects on satiation are no longer apparent after 2 weeks, coinciding with weight gain. CONCLUSIONS: Exposure to obesogenic diets engages homeostatic regulatory mechanisms for ~2 weeks that ultimately fail, and consequent weight gain is characterized by impaired satiation and satiety. Insights into the etiology of obesity can be obtained by investigating changes to satiation and satiety mechanisms during the initial ~2 weeks of HFD exposure.


Sujet(s)
Alimentation riche en graisse , Ration calorique , Métabolisme énergétique , Comportement alimentaire , Hyperphagie , Souris de lignée C57BL , Obésité , Prise de poids , Animaux , Mâle , Souris , Alimentation riche en graisse/effets indésirables , Femelle , Obésité/étiologie , Obésité/métabolisme , Hyperphagie/étiologie , Comportement alimentaire/physiologie , Facteurs temps , Conditionnement physique d'animal , Satiété , Consommation alimentaire/physiologie
8.
Cereb Cortex ; 34(6)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38934712

RÉSUMÉ

In addition to metabolic and cardiovascular disorders, obesity is associated with cognitive deficits in humans and animal models. We have previously shown that obesogenic high-fat and sugar diet intake during adolescence (adoHFSD) impairs hippocampus (HPC)-dependent memory in rodents. These results were obtained in males only and it remains to evaluate whether adoHFSD has similar effect in females. Therefore, here, we investigated the effects of adoHFSD consumption on HPC-dependent contextual fear memory and associated brain activation in male and female mice. Exposure to adoHFSD increased fat mass accumulation and glucose levels in both males and females but impaired contextual fear memory only in males. Compared with females, contextual fear conditioning induced higher neuronal activation in the dorsal and ventral HPC (CA1 and CA3 subfields) as well as in the medial prefrontal cortex in males. Also, adoHFSD-fed males showed enhanced c-Fos expression in the dorsal HPC, particularly in the dentate gyrus, and in the basolateral amygdala compared with the other groups. Finally, chemogenetic inactivation of the dorsal HPC rescued adoHFSD-induced memory deficits in males. Our results suggest that males are more vulnerable to the effects of adoHFSD on HPC-dependent aversive memory than females, due to overactivation of the dorsal HPC.


Sujet(s)
Alimentation riche en graisse , Peur , Hippocampe , Mémoire , Souris de lignée C57BL , Obésité , Caractères sexuels , Animaux , Peur/physiologie , Mâle , Femelle , Hippocampe/métabolisme , Alimentation riche en graisse/effets indésirables , Obésité/métabolisme , Obésité/psychologie , Obésité/physiopathologie , Souris , Mémoire/physiologie , Cortex préfrontal/métabolisme
9.
Front Biosci (Landmark Ed) ; 29(6): 208, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38940030

RÉSUMÉ

Polycystic ovary syndrome (PCOS) is a prevalent reproductive, endocrine, and metabolic disease that affects 5-18% of women worldwide, with a rising incidence. Hyperandrogenemia and insulin resistance are two key pathophysiological factors that contribute to PCOS, both of which contribute to a variety of health issues such as menstrual irregularities, obesity, dysfunctional glucose and lipid homeostasis, infertility, mental disorders, and cardiovascular and cerebrovascular diseases. Despite ongoing studies, the origin and pathogenesis of PCOS remain elusive; there is also a clinical need for simpler, more effective, longer lasting, and more comprehensive treatments for women with PCOS. The gut-fat axis, a critical regulatory route for metabolism, endocrine function, and immune response, has received considerable interest in recent years in the research of the etiology and treatment of metabolic illnesses such as type 2 diabetes mellitus and non-alcoholic fatty liver disease. The latest research in PCOS has revealed significant alterations in the homogeneity and phylogenetic diversity of the gut microbiota. Animal research using fecal microbiota transplantation has confirmed the importance of gut microbiota in regulating insulin sensitivity and sex hormone balance in PCOS. Furthermore, studies have shown a decrease in the volume and/or activity of brown adipose tissue (BAT) in PCOS patients, a change that alters adipokine release, leading to insulin resistance and hyperandrogenemia, aggravating PCOS progression. Given the function of BAT in increasing energy expenditure and alleviating metabolic parameters, efforts to activate BAT or induce browning of white adipose tissue have emerged as possible treatments for PCOS. Recent research has suggested that the gut microbiota can influence BAT creation and activity via metabolites such as short-chain fatty acids and bile acids, as well as the gut-brain axis. Cold exposure, healthy dieting, metformin, bariatric surgery, glucagon-like peptide 1 receptor agonists and melatonin have all been shown in basic and clinical studies to modulate BAT activity by influencing the gut microbiota, demonstrating significant clinical potential. However, more studies into the regulation mechanisms of the gut-BAT axis are required to produce more effective, comfortable, and safe tailored therapeutics for PCOS.


Sujet(s)
Tissu adipeux brun , Microbiome gastro-intestinal , Syndrome des ovaires polykystiques , Syndrome des ovaires polykystiques/microbiologie , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/thérapie , Syndrome des ovaires polykystiques/physiopathologie , Humains , Femelle , Microbiome gastro-intestinal/physiologie , Tissu adipeux brun/métabolisme , Animaux , Insulinorésistance , Transplantation de microbiote fécal , Obésité/microbiologie , Obésité/métabolisme , Obésité/thérapie
10.
Med Microbiol Immunol ; 213(1): 11, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38940844

RÉSUMÉ

BACKGROUND: Obesity-associated male infertility is a common complication of obesity and has been increasing in prevalence. Blautia wexlerae has modulation effects on obesity. However, the action of B. wexlerae on obesity-associated male infertility is unclear. The nod-like receptor protein 3 (NLRP3) inflammasome has become a major target for addressing many diseases, including obesity-associated male infertility. This study aims to investigate the action of B. wexlerae on obesity-associated male infertility and the influence of B. wexlerae on NLRP3 inflammasome. MATERIALS AND METHODS: The fecal samples were collected from 60 infertile men with or without obesity and 30 healthy men. The obesity mice model was established through high-fat diet (HFD) induction. The mating assays evaluated the male infertility of obese mice. A mouse-derived spermatogonia (GC-1 spg) cell viability was detected using the Cell Counting Kit-8 assay. The reactive oxygen species (ROS) were assessed using flow cytometry. Furthermore, immunofluorescence, enzyme-linked immunosorbent assay, and western blotting were applied to measure the gene expressions. RESULTS: Blautia wexlerae was decreased and negatively correlated with interleukin-1 beta (IL-1ß) or IL-18 levels in infertile men with obesity. On the other hand, B. wexlerae improved the mating capability of obese male mice and suppressed oxidative stress and NLRP3 inflammasome via the activation of the acetate receptor. Furthermore, sodium acetate regulated oxidative stress and NLRP3 inflammasome via the activation of the acetate receptor in GC-1 spg cells in vitro. CONCLUSION: The administration of Blautia wexlerae improved obesity-associated male infertility and regulated oxidative stress and NLRP3 inflammasome activities. In general, its administration may be an effective strategy for the treatment of obesity-associated male infertility.


Sujet(s)
Infertilité masculine , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Obésité , Stress oxydatif , Mâle , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Animaux , Obésité/complications , Obésité/métabolisme , Humains , Infertilité masculine/étiologie , Infertilité masculine/métabolisme , Inflammasomes/métabolisme , Souris , Adulte , Espèces réactives de l'oxygène/métabolisme , Modèles animaux de maladie humaine , Alimentation riche en graisse , Interleukine-1 bêta/métabolisme , Souris de lignée C57BL
11.
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
12.
PLoS One ; 19(6): e0305912, 2024.
Article de Anglais | MEDLINE | ID: mdl-38935642

RÉSUMÉ

Diet-induced obesity reduces oocyte quality mainly by impacting oocyte mitochondrial functions. Moreover, maternal obesity is associated with mitochondrial dysfunction in oocytes of their adult offspring. However, these effects were reported only in fully grown oocytes, mainly in the form of abnormal mitochondrial ultrastructure. It is unknown if obesogenic (OB) diets or maternal obesity already impact the primordial and preantral follicles. Considering the long duration and dynamics of folliculogenesis, determining the stage at which oocytes are affected and the extent of the damage is crucial for optimal reproductive management of obese patients and their daughters. Potential interaction between maternal and offspring diet effects are also not described, yet pivotal in our contemporary society. Therefore, here we examined the impact of OB diets on oocyte mitochondrial ultrastructure in primordial and activated preantral follicles in offspring from diet-induced obese or lean mothers. We used an outbred Swiss mouse model to increase the pathophysiological relevance to humans. Female mice were fed control or OB diets for 7 weeks, then mated with control males. Their female offspring were fed control or OB diets after weaning for 7 weeks (2-by-2 factorial design). Adult offspring ovarian sections were examined using transmission electron microscopy. We characterised and classified unique features of oocyte mitochondrial ultrastructure in the preantral follicles. An increase in mitochondrial matrix density was the most predominant change during follicle activation in secondary follicles, a feature that is linked with a higher mitochondrial activity. Maternal obesity increased mitochondrial density already in the primordial follicles suggesting an earlier increase in bioenergetic capacity. Maternal obesity did not induce abberant ultrastructure (abnormalities and defects) in primordial or preantral follicles. In contrast, offspring OB diet increased mitochondrial abnormalities in the primordial follicles. Further investigation of the consequences of these changes on oocyte metabolic regulation and stress levels during folliculogenesis is needed.


Sujet(s)
Mitochondries , Ovocytes , Follicule ovarique , Animaux , Ovocytes/ultrastructure , Ovocytes/métabolisme , Femelle , Follicule ovarique/métabolisme , Follicule ovarique/ultrastructure , Follicule ovarique/anatomopathologie , Souris , Mitochondries/ultrastructure , Mitochondries/métabolisme , Grossesse , Obésité/étiologie , Obésité/anatomopathologie , Obésité/métabolisme , Mâle , Obésité maternelle/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/anatomopathologie , Alimentation riche en graisse/effets indésirables
13.
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
14.
Nat Commun ; 15(1): 5394, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918428

RÉSUMÉ

Adipose tissue macrophages (ATMs) influence obesity-associated metabolic dysfunction, but the mechanisms by which they do so are not well understood. We show that miR-6236 is a bona fide miRNA that is secreted by ATMs during obesity. Global or myeloid cell-specific deletion of miR-6236 aggravates obesity-associated adipose tissue insulin resistance, hyperglycemia, hyperinsulinemia, and hyperlipidemia. miR-6236 augments adipocyte insulin sensitivity by inhibiting translation of negative regulators of insulin signaling, including PTEN. The human genome harbors a miR-6236 homolog that is highly expressed in the serum and adipose tissue of obese people. hsa-MIR-6236 expression negatively correlates with hyperglycemia and glucose intolerance, and positively correlates with insulin sensitivity. Together, our findings establish miR-6236 as an ATM-secreted miRNA that potentiates adipocyte insulin signaling and protects against metabolic dysfunction during obesity.


Sujet(s)
Adipocytes , Hyperglycémie , Insulinorésistance , Insuline , microARN , Obésité , Phosphohydrolase PTEN , Transduction du signal , microARN/métabolisme , microARN/génétique , Obésité/métabolisme , Obésité/génétique , Animaux , Adipocytes/métabolisme , Hyperglycémie/métabolisme , Hyperglycémie/génétique , Humains , Insuline/métabolisme , Insulinorésistance/génétique , Souris , Mâle , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Souris de lignée C57BL , Macrophages/métabolisme , Tissu adipeux/métabolisme , Cellules myéloïdes/métabolisme , Souris knockout , Hyperinsulinisme/métabolisme , Hyperinsulinisme/génétique
15.
Sci Rep ; 14(1): 14685, 2024 06 26.
Article de Anglais | MEDLINE | ID: mdl-38918476

RÉSUMÉ

Human milk oligosaccharides (HMOs) promote adequate intestinal microbiota development and favor the immune system's maturation and cognitive development. In addition to non-modifiable factors, HMOs composition can be influenced by other factors like body mass index and eating habits, but the reports are discrepant. The aim of this work was to describe the correlation between maternal factors and HMOs concentration in colostrum in 70 women from northeastern Mexico categorized into women with normal weight and women with overweight or obesity. The absolute concentration of six HMOs were significantly lower in women with overweight or obesity compared to women with normal weight (LNFPI p = 0.0021, 2'-FL p = 0.0304, LNT p = 0.0492, LNnT p = 0.00026, 3'-SL p = 0.0476, 6'-SL p = 0.00041). Another main finding was that the frequency of consumption of food groups such as vegetables, fruits and meats was positively correlated to specific HMOs (Poblano chili and 2'-FL; rs = 0.702, p = 0.0012; Orange or tangerine and 3-FL; rs = 0.428, p = 0.0022; Chicken and 2'-FL; rs = 0.615, p = 0.0039). This study contributes to the elucidation of how maternal factors influence the composition of HMOs and opens possibilities for future research aimed at mitigating overweight or obesity, consequently improving the quality of human milk.


Sujet(s)
Allaitement naturel , Comportement alimentaire , Lait humain , Oligosaccharides , Humains , Lait humain/composition chimique , Lait humain/métabolisme , Femelle , Mexique , Oligosaccharides/analyse , Adulte , Obésité/métabolisme , Indice de masse corporelle , Colostrum/composition chimique , Colostrum/métabolisme , Surpoids , Jeune adulte
16.
J Transl Med ; 22(1): 592, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918843

RÉSUMÉ

BACKGROUND: Fundamentally defined by an imbalance in energy consumption and energy expenditure, obesity is a significant risk factor of several musculoskeletal conditions including osteoarthritis (OA). High-fat diets and sedentary lifestyle leads to increased adiposity resulting in systemic inflammation due to the endocrine properties of adipose tissue producing inflammatory cytokines and adipokines. We previously showed serum levels of specific adipokines are associated with biomarkers of bone remodelling and cartilage volume loss in knee OA patients. Whilst more recently we find the metabolic consequence of obesity drives the enrichment of pro-inflammatory fibroblast subsets within joint synovial tissues in obese individuals compared to those of BMI defined 'health weight'. As such this present study identifies obesity-associated genes in OA joint tissues which are conserved across species and conditions. METHODS: The study utilised 6 publicly available bulk and single-cell transcriptomic datasets from human and mice studies downloaded from Gene Expression Omnibus (GEO). Machine learning models were employed to model and statistically test datasets for conserved gene expression profiles. Identified genes were validated in OA tissues from obese and healthy weight individuals using quantitative PCR method (N = 38). Obese and healthy-weight patients were categorised by BMI > 30 and BMI between 18 and 24.9 respectively. Informed consent was obtained from all study participants who were scheduled to undergo elective arthroplasty. RESULTS: Principal component analysis (PCA) was used to investigate the variations between classes of mouse and human data which confirmed variation between obese and healthy populations. Differential gene expression analysis filtered on adjusted p-values of p < 0.05, identified differentially expressed genes (DEGs) in mouse and human datasets. DEGs were analysed further using area under curve (AUC) which identified 12 genes. Pathway enrichment analysis suggests these genes were involved in the biosynthesis and elongation of fatty acids and the transport, oxidation, and catabolic processing of lipids. qPCR validation found the majority of genes showed a tendency to be upregulated in joint tissues from obese participants. Three validated genes, IGFBP2 (p = 0.0363), DOK6 (0.0451) and CASP1 (0.0412) were found to be significantly different in obese joint tissues compared to lean-weight joint tissues. CONCLUSIONS: The present study has employed machine learning models across several published obesity datasets to identify obesity-associated genes which are validated in joint tissues from OA. These results suggest obesity-associated genes are conserved across conditions and may be fundamental in accelerating disease in obese individuals. Whilst further validations and additional conditions remain to be tested in this model, identifying obesity-associated genes in this way may serve as a global aid for patient stratification giving rise to the potential of targeted therapeutic interventions in such patient subpopulations.


Sujet(s)
Obésité , Transcriptome , Humains , Obésité/génétique , Obésité/complications , Obésité/métabolisme , Animaux , Souris , Transcriptome/génétique , Spécificité d'espèce , Analyse de profil d'expression de gènes , Analyse en composantes principales , Apprentissage machine , Régulation de l'expression des gènes , Mâle , Femelle
17.
Cancer Med ; 13(12): e7346, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923758

RÉSUMÉ

OBJECTIVES: Previous studies have demonstrated that obesity may impact the efficacy of anti-PD1 therapy, but the underlying mechanism remains unclear. In this study, our objective was to determine the prognostic value of obesity in patients with oral tongue squamous cell carcinoma (OTSCC) treated with pembrolizumab and establish a subtype based on fatty acid metabolism-related genes (FAMRGs) for immunotherapy. MATERIALS AND METHODS: We enrolled a total of 56 patients with OTSCC who underwent neoadjuvant anti-PD1 therapy. Univariate and multivariate Cox regression analyses, Kaplan-Meier survival analysis, and immunohistochemistry staining were performed. Additionally, we acquired the gene expression profiles of pan-cancer samples and conducted GSEA and KEGG pathway analysis. Moreover, data from TCGA, MSigDB, UALCAN, GEPIA and TIMER were utilized to construct the FAMRGs subtype. RESULTS: Our findings indicate that high Body Mass Index (BMI) was significantly associated with improved PFS (HR = 0.015; 95% CI, 0.001 to 0.477; p = 0.015), potentially attributed to increased infiltration of PD1 + T cells. A total of 91 differentially expressed FAMRGs were identified between the response and non-response groups in pan-cancer patients treated with immunotherapy. Of these, 6 hub FAMRGs (ACSL5, PLA2G2D, PROCA1, IL4I1, UBE2L6 and PSME1) were found to affect PD-1 expression and T cell infiltration in HNSCC, which may impact the efficacy of anti-PD1 therapy. CONCLUSION: This study demonstrates that obesity serves as a robust prognostic predictor for patients with OTSCC undergoing neoadjuvant anti-PD1 therapy. Furthermore, the expression of 6 hub FAMRGs (ACSL5, PLA2G2D, PROCA1, IL4I1, UBE2L6 and PSME1) plays a pivotal role in the context of anti-PD1 therapy and deserves further investigation.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Traitement néoadjuvant , Obésité , Tumeurs de la langue , Humains , Tumeurs de la langue/traitement médicamenteux , Tumeurs de la langue/métabolisme , Tumeurs de la langue/immunologie , Tumeurs de la langue/anatomopathologie , Tumeurs de la langue/mortalité , Tumeurs de la langue/génétique , Femelle , Mâle , Traitement néoadjuvant/méthodes , Obésité/métabolisme , Obésité/complications , Adulte d'âge moyen , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Pronostic , Sujet âgé , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/mortalité , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/thérapie , Anticorps monoclonaux humanisés/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Adulte , Indice de masse corporelle , Marqueurs biologiques tumoraux/métabolisme
18.
PLoS One ; 19(6): e0306038, 2024.
Article de Anglais | MEDLINE | ID: mdl-38924041

RÉSUMÉ

BACKGROUND: The dual existence of Type 2 Diabetes Mellitus (T2DM) and obesity within a single individual may describe a combined adverse health effects, including impaired quality of life and increased risk for cardiovascular diseases (CVDs). Oxidative stress is a contributing factor to the pathogenesis of obesity. Meanwhile, dietary antioxidants may improve the antioxidant defense system and thereby decrease oxidative injury. Dietary total antioxidant capacity (TAC) is usually used to investigate the potential health effects of dietary antioxidant intake on several oxidative stress induced chronic diseases. This study aimed to examine the association of dietary TAC with obesity-related features in T2DM patients. METHODS: The present study included 254 type 2 diabetes outpatients with a mean (SD) age of 54.52 (7.21) years and mean (SD) diabetes duration of 8.2 (6.4) years. Data on dietary intake was assessed using a validated food frequency questionnaire. Dietary TAC was estimated by ferric reducing antioxidant potential (FRAP) method. Anthropometric, clinical and lifestyle characteristics were all collected. RESULTS: In linear regression analyses, dietary antioxidant capacity was inversely associated with body mass index (ß = -0,231; 95% CI, -0,419 to -0,042), waist circumference (ß = -0,427; 95% CI, -0,849 to -0,006) and fat mass percentage (ß = -0,328; 95% CI, -0,545 to -0,112) independently of the assessed confounding variables. Interestingly, dietary TAC showed positive and significant associations with vitamin A, vitamin C, ß-carotene, magnesium, folic acid and iron intakes, after adjusting for age and daily energy intake. CONCLUSIONS: Higher intake of dietary TAC was in association with lower indices of general and central obesity in T2DM patients. Therefore, dietary recommendations for counteracting obesity in patients with T2DM should take into account a high dietary TAC.


Sujet(s)
Antioxydants , Diabète de type 2 , Obésité abdominale , Humains , Diabète de type 2/diétothérapie , Diabète de type 2/complications , Diabète de type 2/métabolisme , Adulte d'âge moyen , Antioxydants/métabolisme , Antioxydants/administration et posologie , Femelle , Mâle , Obésité abdominale/complications , Régime alimentaire , Indice de masse corporelle , Stress oxydatif/effets des médicaments et des substances chimiques , Obésité/métabolisme , Obésité/complications , Obésité/diétothérapie , Tour de taille , Adulte
19.
Sci Adv ; 10(26): eadn5229, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38924414

RÉSUMÉ

There is a regional preference around lymph nodes (LNs) for adipose beiging. Here, we show that local LN removal within inguinal white adipose tissue (iWAT) greatly impairs cold-induced beiging, and this impairment can be restored by injecting M2 macrophages or macrophage-derived C-C motif chemokine (CCL22) into iWAT. CCL22 injection into iWAT effectively promotes iWAT beiging, while blocking CCL22 with antibodies can prevent it. Mechanistically, the CCL22 receptor, C-C motif chemokine receptor 4 (CCR4), within eosinophils and its downstream focal adhesion kinase/p65/interleukin-4 signaling are essential for CCL22-mediated beige adipocyte formation. Moreover, CCL22 levels are inversely correlated with body weight and fat mass in mice and humans. Acute elevation of CCL22 levels effectively prevents diet-induced body weight and fat gain by enhancing adipose beiging. Together, our data identify the CCL22-CCR4 axis as an essential mediator for LN-controlled adaptive thermogenesis and highlight its potential to combat obesity and its associated complications.


Sujet(s)
Tissu adipeux blanc , Chimiokine CCL22 , Métabolisme énergétique , Noeuds lymphatiques , Macrophages , Thermogenèse , Chimiokine CCL22/métabolisme , Animaux , Macrophages/métabolisme , Souris , Humains , Noeuds lymphatiques/métabolisme , Tissu adipeux blanc/métabolisme , Mâle , Récepteurs CCR4/métabolisme , Obésité/métabolisme , Transduction du signal , Souris de lignée C57BL , Granulocytes éosinophiles/métabolisme , Femelle , Adipocytes beiges/métabolisme
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...