Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 18.279
Filtrer
1.
Lab Anim (NY) ; 53(7): 172, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38956364
2.
Cell Metab ; 36(7): 1494-1503.e3, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38959863

RÉSUMÉ

The extent to which modifiable lifestyle factors offset the determined genetic risk of obesity and obesity-related morbidities remains unknown. We explored how the interaction between genetic and lifestyle factors influences the risk of obesity and obesity-related morbidities. The polygenic score for body mass index was calculated to quantify inherited susceptibility to obesity in 338,645 UK Biobank European participants, and a composite lifestyle score was derived from five obesogenic factors (physical activity, diet, sedentary behavior, alcohol consumption, and sleep duration). We observed significant interaction between high genetic risk and poor lifestyles (pinteraction < 0.001). Absolute differences in obesity risk between those who adhere to healthy lifestyles and those who do not had gradually expanded with an increase in polygenic score. Despite a high genetic risk for obesity, individuals can prevent obesity-related morbidities by adhering to a healthy lifestyle and maintaining a normal body weight. Healthy lifestyles should be promoted irrespective of genetic background.


Sujet(s)
Indice de masse corporelle , Prédisposition génétique à une maladie , Mode de vie , Obésité , Humains , Obésité/génétique , Mâle , Femelle , Adulte d'âge moyen , Facteurs de risque , Adulte , Sujet âgé , Exercice physique , Mode de vie sédentaire , Royaume-Uni/épidémiologie
3.
Biol Direct ; 19(1): 52, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956667

RÉSUMÉ

BACKGROUND: Adiposity profoundly impacts reproductive health in both humans and animals. However, the precise subpopulations contributing to infertility under obese conditions remain elusive. RESULTS: In this study, we established an obese mouse model through an eighteen-week high-fat diet regimen in adult female mice. Employing single-cell RNA sequencing (scRNA-seq), we constructed a comprehensive single-cell atlas of ovarian tissues from these mice to scrutinize the impact of obesity on the ovarian microenvironment. ScRNA-seq revealed notable alterations in the microenvironment of ovarian tissues in obese mice. Granulosa cells, stromal cells, T cells, and macrophages exhibited functional imbalances compared to the control group. We observed heightened interaction strength in the SPP1-CD44 pairing within lgfbp7+ granulosa cell subtypes and Il1bhigh monocyte subtypes in the ovarian tissues of obese mice. Moreover, the interaction strength between Il1bhigh monocyte subtypes and Pdgfrb+ stromal cell subtypes in the form of TNF - TNFrsf1α interaction was also enhanced subsequently to obesity, potentially contributing to ovarian fibrosis pathogenesis. CONCLUSIONS: We propose a model wherein granulosa cells secrete SPP1 to activate monocytes, subsequently triggering TNF-α secretion by monocytes, thereby activating stromal cells and ultimately leading to the development of ovarian fibrosis. Intervening in this process may represent a promising avenue for improving clinical outcomes in fertility treatments for obese women.


Sujet(s)
Fibrose , Souris obèse , Obésité , Analyse sur cellule unique , Animaux , Femelle , Souris , Fibrose/génétique , Obésité/génétique , Obésité/métabolisme , Analyse de profil d'expression de gènes , Ovaire/métabolisme , Transcriptome , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables , Cellules de la granulosa/métabolisme
4.
Front Endocrinol (Lausanne) ; 15: 1379293, 2024.
Article de Anglais | MEDLINE | ID: mdl-38978626

RÉSUMÉ

Background: Polycystic ovary syndrome (PCOS) is the most common reproductive-endocrine disorder with wide-ranging metabolic implications, including obesity. RNA editing, a post-transcriptional modification, can fine-tune protein function and introduce heterogeneity. However, the role of RNA editing and its impact on adipose tissue function in PCOS remain poorly understood. Methods: This study aimed to comprehensively analyze RNA-editing events in abdominal and subcutaneous adipose tissue of PCOS patients and healthy controls using high-throughput whole-genome sequencing (WGS) and RNA sequencing. Results: Our results revealed that PCOS patients exhibited more RNA-editing sites, with adenosine-to-inosine (A-to-I) editing being prevalent. The expression of ADAR genes, responsible for A-to-I editing, was also higher in PCOS. Aberrant RNA-editing sites in PCOS adipose tissue was enriched in immune responses, and interleukin-12 biosynthetic process. Tumor necrosis factor (TNF) signaling, nuclear factor kappa B (NF-κB) signaling, Notch signaling, terminal uridylyl transferase 4 (TUT4), hook microtubule tethering protein 3 (HOOK3), and forkhead box O1 (FOXO1) were identified to be of significant differences. Differentially expressed genes (DEGs) in PCOS adipose tissue were enriched in immune responses compared with controls, and the DEGs between subcutaneous and abdominal adipose tissue were also enriched in immune responses suggesting the important role of subcutaneous adipose tissue. Furthermore, we identified the correlations between RNA editing levels and RNA expression levels of specific genes, such as ataxia-telangiectasia mutated (ATM) and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) in inflammation pathways and ATM, TUT4, and YTH N6-methyladenosine RNA-binding protein C2 (YTHDC2) in oocyte development pathway. Conclusions: These findings suggest that RNA-editing dysregulation in PCOS adipose tissue may contribute to inflammatory dysregulations. Understanding the interplay between RNA editing and adipose tissue function may unveil potential therapeutic targets for PCOS management. However, further research and validation are required to fully elucidate the molecular mechanisms underlying these associations.


Sujet(s)
Tissu adipeux , Obésité , Syndrome des ovaires polykystiques , Édition des ARN , Humains , Syndrome des ovaires polykystiques/génétique , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/immunologie , Syndrome des ovaires polykystiques/anatomopathologie , Femelle , Obésité/génétique , Obésité/métabolisme , Adulte , Tissu adipeux/métabolisme , Études cas-témoins , Séquençage du génome entier
5.
FASEB J ; 38(13): e23794, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38967258

RÉSUMÉ

Obesity is often associated with low-grade inflammation. The incidence of obesity has increased annually worldwide, which seriously affects human health. A previous study indicated that long noncoding RNA SNHG12 was downregulated in obesity. Nevertheless, the role of SNHG12 in obesity remains to be elucidated. In this study, qRT-PCR, western blot, and ELISA were utilized to examine the gene and protein expression. Flow cytometry was employed to investigate the M2 macrophage markers. RNA pull-down assay and RIP were utilized to confirm the interactions of SNHG12, hnRNPA1, and HDAC9. Eventually, a high-fat diet-fed mouse model was established for in vivo studies. SNHG12 overexpression suppressed adipocyte inflammation and insulin resistance and promoted M2 polarization of macrophages that was caused by TNF-α treatment. SNHG12 interacted with hnRNPA1 to downregulate HDAC9 expression, which activated the Nrf2 signaling pathway. HDAC9 overexpression reversed the effect of SNHG12 overexpression on inflammatory response, insulin resistance, and M2 phenotype polarization. Overexpression of SNHG12 improved high-fat diet-fed mouse tissue inflammation. This study revealed the protective effect of SNHG12 against adipocyte inflammation and insulin resistance. This result further provides a new therapeutic target for preventing inflammation and insulin resistance in obesity.


Sujet(s)
Adipocytes , Alimentation riche en graisse , Histone deacetylases , Inflammation , Insulinorésistance , Souris de lignée C57BL , Facteur-2 apparenté à NF-E2 , Obésité , ARN long non codant , Protéines de répression , Animaux , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris , Inflammation/métabolisme , Inflammation/génétique , Adipocytes/métabolisme , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Alimentation riche en graisse/effets indésirables , Mâle , Obésité/métabolisme , Obésité/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Transduction du signal , Macrophages/métabolisme
6.
Front Public Health ; 12: 1366838, 2024.
Article de Anglais | MEDLINE | ID: mdl-38947357

RÉSUMÉ

Background: In recent years, the prevalence of obesity has continued to increase as a global health concern. Numerous epidemiological studies have confirmed the long-term effects of exposure to ambient air pollutant particulate matter 2.5 (PM2.5) on obesity, but their relationship remains ambiguous. Methods: Utilizing large-scale publicly available genome-wide association studies (GWAS), we conducted univariate and multivariate Mendelian randomization (MR) analyses to assess the causal effect of PM2.5 exposure on obesity and its related indicators. The primary outcome given for both univariate MR (UVMR) and multivariate MR (MVMR) is the estimation utilizing the inverse variance weighted (IVW) method. The weighted median, MR-Egger, and maximum likelihood techniques were employed for UVMR, while the MVMR-Lasso method was applied for MVMR in the supplementary analyses. In addition, we conducted a series of thorough sensitivity studies to determine the accuracy of our MR findings. Results: The UVMR analysis demonstrated a significant association between PM2.5 exposure and an increased risk of obesity, as indicated by the IVW model (odds ratio [OR]: 6.427; 95% confidence interval [CI]: 1.881-21.968; P FDR = 0.005). Additionally, PM2.5 concentrations were positively associated with fat distribution metrics, including visceral adipose tissue (VAT) (OR: 1.861; 95% CI: 1.244-2.776; P FDR = 0.004), particularly pancreatic fat (OR: 3.499; 95% CI: 2.092-5.855; PFDR =1.28E-05), and abdominal subcutaneous adipose tissue (ASAT) volume (OR: 1.773; 95% CI: 1.106-2.841; P FDR = 0.019). Furthermore, PM2.5 exposure correlated positively with markers of glucose and lipid metabolism, specifically triglycerides (TG) (OR: 19.959; 95% CI: 1.269-3.022; P FDR = 0.004) and glycated hemoglobin (HbA1c) (OR: 2.462; 95% CI: 1.34-4.649; P FDR = 0.007). Finally, a significant negative association was observed between PM2.5 concentrations and levels of the novel obesity-related biomarker fibroblast growth factor 21 (FGF-21) (OR: 0.148; 95% CI: 0.025-0.89; P FDR = 0.037). After adjusting for confounding factors, including external smoke exposure, physical activity, educational attainment (EA), participation in sports clubs or gym leisure activities, and Townsend deprivation index at recruitment (TDI), the MVMR analysis revealed that PM2.5 levels maintained significant associations with pancreatic fat, HbA1c, and FGF-21. Conclusion: Our MR study demonstrates conclusively that higher PM2.5 concentrations are associated with an increased risk of obesity-related indicators such as pancreatic fat content, HbA1c, and FGF-21. The potential mechanisms require additional investigation.


Sujet(s)
Étude d'association pangénomique , Analyse de randomisation mendélienne , Obésité , Matière particulaire , , Humains , Obésité/génétique , /génétique , Polluants atmosphériques/effets indésirables , Exposition environnementale/effets indésirables , Pollution de l'air/effets indésirables
7.
J Biochem Mol Toxicol ; 38(7): e23764, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38963172

RÉSUMÉ

Obesity is an established risk factor for numerous malignancies, although it remains uncertain whether the disease itself or weight-loss drugs are responsible for a greater predisposition to cancer. The objective of the current study was to determine the impact of dulaglutide on genetic and epigenetic DNA damage caused by obesity, which is a crucial factor in the development of cancer. Mice were administered a low-fat or high-fat diet for 12 weeks, followed by a 5-week treatment with dulaglutide. Following that, modifications of the DNA bases were examined using the comet assay. To clarify the underlying molecular mechanisms, oxidized and methylated DNA bases, changes in the redox status, levels of inflammatory cytokines, and the expression levels of some DNA repair genes were evaluated. Animals fed a high-fat diet exhibited increased body weights, elevated DNA damage, oxidation of DNA bases, and DNA hypermethylation. In addition, obese mice showed altered inflammatory responses, redox imbalances, and repair gene expressions. The findings demonstrated that dulaglutide does not exhibit genotoxicity in the investigated conditions. Following dulaglutide administration, animals fed a high-fat diet demonstrated low DNA damage, less oxidation and methylation of DNA bases, restored redox balance, and improved inflammatory responses. In addition, dulaglutide treatment restored the upregulated DNMT1, Ogg1, and p53 gene expression. Overall, dulaglutide effectively maintains DNA integrity in obese animals. It reduces oxidative DNA damage and hypermethylation by restoring redox balance, modulating inflammatory responses, and recovering altered gene expressions. These findings demonstrate dulaglutide's expediency in treating obesity and its associated complications.


Sujet(s)
Altération de l'ADN , Méthylation de l'ADN , Réparation de l'ADN , Alimentation riche en graisse , Peptides glucagon-like , Fragments Fc des immunoglobulines , Oxydoréduction , Protéines de fusion recombinantes , Animaux , Peptides glucagon-like/analogues et dérivés , Peptides glucagon-like/pharmacologie , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Fragments Fc des immunoglobulines/pharmacologie , Altération de l'ADN/effets des médicaments et des substances chimiques , Souris , Réparation de l'ADN/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Protéines de fusion recombinantes/pharmacologie , Mâle , Oxydoréduction/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/génétique , Stress oxydatif/effets des médicaments et des substances chimiques , Obésité/métabolisme , Obésité/traitement médicamenteux , Obésité/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Souris de lignée C57BL
8.
Front Endocrinol (Lausanne) ; 15: 1385463, 2024.
Article de Anglais | MEDLINE | ID: mdl-38974580

RÉSUMÉ

Melanocortin 4 receptor (MC4R) mutations are the commonest cause of monogenic obesity through dysregulation of neuronal pathways in the hypothalamus and prefrontal cortex that regulate hunger and satiety. MC4R also regulates neuropathic pain pathways via JNK signaling after nerve injury. We show evidence of corneal small fiber degeneration in 2 siblings carrying a heterozygous missense variant c.508A>G, p.Ille170Val in the MC4R gene. Both children were treated with once weekly semaglutide for 6 months with no change in weight, and only a minor improvement in HbA1c and lipid profile. However, there was evidence of nerve regeneration with an increase in corneal nerve fiber density (CNFD) [child A (13.9%), child B (14.7%)], corneal nerve branch density (CNBD) [child A (110.2%), child B (58.7%)] and corneal nerve fiber length (CNFL) [child A (21.5%), child B (44.0%)].


Sujet(s)
Régénération nerveuse , Récepteur de la mélanocortine de type 4 , Humains , Récepteur de la mélanocortine de type 4/génétique , Mâle , Femelle , Enfant , Régénération nerveuse/effets des médicaments et des substances chimiques , Peptides glucagon-like/usage thérapeutique , Peptides glucagon-like/pharmacologie , Neurofibres/effets des médicaments et des substances chimiques , Neurofibres/anatomopathologie , Mutation , Obésité/traitement médicamenteux , Obésité/génétique , Cornée/effets des médicaments et des substances chimiques , Cornée/innervation , Cornée/anatomopathologie , Obésité pédiatrique/traitement médicamenteux , Adolescent
9.
JCI Insight ; 9(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38973609

RÉSUMÉ

Lipoprotein lipase (LPL) hydrolyzes circulating triglycerides (TGs), releasing fatty acids (FA) and promoting lipid storage in white adipose tissue (WAT). However, the mechanisms regulating adipose LPL and its relationship with the development of hypertriglyceridemia are largely unknown. WAT from obese humans exhibited high PAR2 expression, which was inversely correlated with the LPL gene. Decreased LPL expression was also inversely correlated with elevated plasma TG levels, suggesting that adipose PAR2 might regulate hypertriglyceridemia by downregulating LPL. In mice, aging and high palmitic acid diet (PD) increased PAR2 expression in WAT, which was associated with a high level of macrophage migration inhibitory factor (MIF). MIF downregulated LPL expression and activity in adipocytes by binding with CXCR2/4 receptors and inhibiting Akt phosphorylation. In a MIF overexpression model, high-circulating MIF levels suppressed adipose LPL, and this suppression was associated with increased plasma TGs but not FA. Following PD feeding, adipose LPL expression and activity were significantly reduced, and this reduction was reversed in Par2-/- mice. Recombinant MIF infusion restored high plasma MIF levels in Par2-/- mice, and the levels decreased LPL and attenuated adipocyte lipid storage, leading to hypertriglyceridemia. These data collectively suggest that downregulation of adipose LPL by PAR2/MIF may contribute to the development of hypertriglyceridemia.


Sujet(s)
Régulation négative , Hypertriglycéridémie , Lipoprotein lipase , Récepteur de type PAR-2 , Animaux , Lipoprotein lipase/métabolisme , Lipoprotein lipase/génétique , Hypertriglycéridémie/métabolisme , Hypertriglycéridémie/génétique , Souris , Humains , Récepteur de type PAR-2/métabolisme , Récepteur de type PAR-2/génétique , Mâle , Souris knockout , Triglycéride/métabolisme , Triglycéride/sang , Tissu adipeux blanc/métabolisme , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Facteurs inhibiteurs de la migration des macrophages/génétique , Adipocytes/métabolisme , Obésité/métabolisme , Obésité/génétique , Acide palmitique/métabolisme , Femelle , Souris de lignée C57BL , Adulte d'âge moyen
10.
Sci Rep ; 14(1): 15717, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977823

RÉSUMÉ

Obesity is a global health concern and independent risk factor for cancers including hepatocellular carcinoma (HCC). However, evidence on the causal links between obesity and HCC is limited and inconclusive. This study aimed to investigate the causal relationship between obesity-related traits and HCC risk and explore underlying mechanisms using bioinformatics approaches. Two-sample Mendelian randomization analysis was conducted leveraging publicly available genome-wide association study summary data on obesity traits (body mass index, body fat percentage, waist circumference, waist-to-hip ratio, visceral adipose tissue volume) and HCC. Associations of obesity with primary mechanisms (insulin resistance, adipokines, inflammation) and their effects on HCC were examined. Differentially expressed genes in obesity and HCC were identified and functional enrichment analyses were performed. Correlations with tumor microenvironment (TME) and immunotherapy markers were analyzed. Genetically predicted higher body mass index and body fat percentage showed significant causal relationships with increased HCC risk. Overall obesity also demonstrated causal links with insulin resistance, circulating leptin levels, C-reactive protein levels and risk of severe insulin resistant type 2 diabetes. Four differentially expressed genes (ESR1, GCDH, FAHD2A, DCXR) were common in obesity and HCC. Enrichment analyses indicated their roles in processes like RNA capping, viral transcription, IL-17 signaling and endocrine resistance. They exhibited negative correlations with immune cell infiltration and immunotherapy markers in HCC. Overall obesity likely has a causal effect on HCC risk in Europeans, possibly via influencing primary mechanisms. The identified differentially expressed genes may be implicated in obesity-induced hepatocarcinogenesis through regulating cell cycle, inflammation and immune evasion. Further research on precise mechanisms is warranted.


Sujet(s)
Carcinome hépatocellulaire , Étude d'association pangénomique , Tumeurs du foie , Obésité , Humains , Carcinome hépatocellulaire/génétique , Tumeurs du foie/génétique , Obésité/complications , Obésité/génétique , Indice de masse corporelle , Facteurs de risque , Insulinorésistance/génétique , Microenvironnement tumoral/génétique , Analyse de randomisation mendélienne
11.
Physiol Rep ; 12(13): e16140, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38997217

RÉSUMÉ

The brain derived-neurotrophic factor (BDNF) Val66Met polymorphism causes functional changes in BDNF, and is associated with obesity and some psychiatric disorders, but its relationship to health-related quality of life (HRQoL) remains unknown. This study examined, in youth with obesity, whether carriers of the BDNF Val66met polymorphism Met-alleles (A/A or G/A) differed from noncarriers (G/G) on HRQoL. The participants were 187 adolescents with obesity. Ninety-nine youth were carriers of the homozygous Val/Val (G/G) alleles, and 88 were carriers of the Val/Met (G/A) or Met/Met (A/A) alleles. Blood samples were drawn in the morning after an overnight fast for genotyping. HRQoL was measured using the Pediatric-Quality of Life core version. Compared to carriers of the Val66Met Val (G/G) alleles, carriers of the Met-Alleles reported significantly higher physical -HRQoL (p = 0.02), school-related HRQoL, (p = 0.05), social-related HRQoL (p = 0.05), and total HRQoL (p = 0.03), and a trend for Psychosocial-HRQoL. Research is needed to confirm our findings and determine whether carriers of the BDNF Val66Met homozygous Val (G/G) alleles may be at risk of diminished HRQoL, information that can influence interventions in a high-risk population of inactive youth with obesity.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Polymorphisme de nucléotide simple , Qualité de vie , Humains , Facteur neurotrophique dérivé du cerveau/génétique , Facteur neurotrophique dérivé du cerveau/sang , Mâle , Adolescent , Femelle , Enfant , Obésité/génétique , Obésité/psychologie , Obésité pédiatrique/génétique , Obésité pédiatrique/psychologie
12.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38999965

RÉSUMÉ

Female infertility constitutes a growing health problem in developing countries and could be associated with several possible causes including reproductive disorders, congenital malformations, infections and hormonal dysfunction. Nonetheless, a series of additional factors can also negatively impact female fertility and are represented by chronic exposure to environmental pollutants, stress, unhealthy lifestyle choices such as cigarette smoking and, among others, obesity. Excess weight is associated with several chronic diseases, and growing evidence demonstrates that it can compromise reproductive physiology due to its influence on endometrial gene expression and receptivity. Thus, the current review of the literature mainly focused on how obesity can impair uterine receptivity, mostly from a molecular point of view throughout the window of implantation (WOI) period at an endometrial level. It was also highlighted that an obesity-related increase in adipose tissue may lead to a modulation in the expression of multiple pathways, which could cause a hostile endometrial environment with a consequent negative impact on the uterine receptivity and the establishment of pregnancy. Thanks to the use of the endometrial receptivity assay (ERA), a specific microarray that studies the expression of a series of genes, it is now possible to evaluate the endometrial status of patients with infertility problems in a more detailed manner. Moreover, female fertility and endometrial receptivity could be affected by endometriosis, a chronic benign gynecological disease, whose cause-and-effect relationship to obesity is still uncertain. Therefore, further investigations would be required to better elucidate these mechanisms that govern embryo implantation and could be potentially useful for the generation of new strategies to overcome implantation failure and improve the pregnancy rates in obese women.


Sujet(s)
Endomètre , Infertilité féminine , Obésité , Humains , Femelle , Obésité/métabolisme , Obésité/génétique , Infertilité féminine/métabolisme , Infertilité féminine/étiologie , Infertilité féminine/génétique , Endomètre/métabolisme , Grossesse , Implantation embryonnaire , Endométriose/métabolisme , Endométriose/génétique , Endométriose/anatomopathologie , Animaux
13.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000282

RÉSUMÉ

Obesity is a global health challenge that has received increasing attention in contemporary research. The gut microbiota has been implicated in the development of obesity, primarily through its involvement in regulating various host metabolic processes. Recent research suggests that epigenetic modifications may serve as crucial pathways through which the gut microbiota and its metabolites contribute to the pathogenesis of obesity and other metabolic disorders. Hence, understanding the interplay between gut microbiota and epigenetic mechanisms is crucial for elucidating the impact of obesity on the host. This review primarily focuses on the understanding of the relationship between the gut microbiota and its metabolites with epigenetic mechanisms in several obesity-related pathogenic mechanisms, including energy dysregulation, metabolic inflammation, and maternal inheritance. These findings could serve as novel therapeutic targets for probiotics, prebiotics, and fecal microbiota transplantation tools in treating metabolic disruptions. It may also aid in developing therapeutic strategies that modulate the gut microbiota, thereby regulating the metabolic characteristics of obesity.


Sujet(s)
Épigenèse génétique , Microbiome gastro-intestinal , Obésité , Humains , Obésité/métabolisme , Obésité/microbiologie , Obésité/génétique , Animaux , Probiotiques , Transplantation de microbiote fécal , Prébiotiques , Métabolisme énergétique
14.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000297

RÉSUMÉ

Obesity is a major public health concern that is associated with negative health outcomes. Exercise and dietary restriction are commonly recommended to prevent or combat obesity. This study investigates how voluntary exercise mitigates abnormal gene expression in the hypothalamic arcuate nucleus (ARC) of diet-induced obese (DIO) rats. Using a transcriptomic approach, novel genes in the ARC affected by voluntary wheel running were assessed alongside physiology, pharmacology, and bioinformatics analysis to evaluate the role of miR-211 in reversing obesity. Exercise curbed weight gain and fat mass, and restored ARC gene expression. High-fat diet (HFD) consumption can dysregulate satiety/hunger mechanisms in the ARC. Transcriptional clusters revealed that running altered gene expression patterns, including inflammation and cellular structure genes. To uncover regulatory mechanisms governing gene expression in DIO attenuation, we explored miR-211, which is implicated in systemic inflammation. Exercise ameliorated DIO overexpression of miR-211, demonstrating its pivotal role in regulating inflammation in the ARC. Further, in vivo central administration of miR-211-mimic affected the expression of immunity and cell cycle-related genes. By cross-referencing exercise-affected and miR-211-regulated genes, potential candidates for obesity reduction through exercise were identified. This research suggests that exercise may rescue obesity through gene expression changes mediated partially through miR-211.


Sujet(s)
Noyau arqué de l'hypothalamus , Alimentation riche en graisse , microARN , Obésité , Conditionnement physique d'animal , Animaux , Noyau arqué de l'hypothalamus/métabolisme , microARN/génétique , microARN/métabolisme , Obésité/génétique , Obésité/métabolisme , Rats , Femelle , Alimentation riche en graisse/effets indésirables , Régulation de l'expression des gènes , Inflammation/génétique , Inflammation/métabolisme
15.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-39000422

RÉSUMÉ

Maternal obesity, caused by diets rich in fats and sugars during pregnancy, can predispose offspring to metabolic diseases such as diabetes. We hypothesized that obesity during pregnancy leads to increased DNA methylation and reduced protein expression in factors regulating ß-cell function and apoptosis. Female C57BL/6J mice were fed a high-fat diet (HFD; 42% fat content; n = 3) or a control diet (CON; 16% fat content; n = 3) for fourteen weeks before and during pregnancy. Offspring were euthanized at 8 weeks and pancreatic tissue was collected. Isolated DNA was analyzed using whole-genome bisulfite sequencing. Protein expression was quantified using LC-MS. No significant differences in body weight were observed between HFD and control pups (p = 0.10). Whole-genome bisulfite sequencing identified 91,703 and 88,415 differentially methylated regions (DMRs) in CON vs. HFD male and female offspring. A total of 34 and 4 proteins were determined to have changes in expression that correlated with changes in DNA methylation in CON vs. HFD males and females, respectively. The majority of these factors were grouped into the metabolic function category via pathway analyses. This study illustrates the complex relationship between epigenetics, diet, and sex-specific responses, therefore offering insights into potential therapeutic targets and areas for further research.


Sujet(s)
Méthylation de l'ADN , Alimentation riche en graisse , Souris de lignée C57BL , Pancréas , Animaux , Femelle , Alimentation riche en graisse/effets indésirables , Grossesse , Souris , Mâle , Pancréas/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/génétique , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Obésité/métabolisme , Obésité/génétique , Obésité/étiologie , Épigenèse génétique , Multi-omique
16.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000558

RÉSUMÉ

Male reproductive dysfunction is a clinical disease, with a large number of cases being idiopathic. Reproductive disorders have been found in obese (diet-induced obesity and diet-induced obesity-resistant) mice, but the mechanism behind the male reproductive dysfunction between them may be different. The purpose of this study was to explore the possible role and mechanism of miR-34c on sperm production in high-fat-diet-induced obesity-resistant (DIO-R) mice and GC-1 spg cells, which may differ from those in high-fat-diet-induced obesity (DIO) mice. In vivo and in vitro experiments were performed. C57BL/6J mice were fed a high-fat diet for 10 weeks to establish the DIO and DIO-R mouse model. GC-1 spg cells were used to verify the mechanism of miR-34c on sperm production. During in vivo experiments, sperm production damage was found in both DIO and DIO-R male mice. Compared to the control mice, significantly decreased levels of testosterone, LH, activities of acrosome enzyme (ACE), HAse, and activating transcription factor 1 (ATF1) were found in both DIO and DIO-R male mice (p < 0.05). Compared with the control group, the ratio of B-cell lymphoma-2 (Bcl-2)/bcl-2-associated X protein (Bax) in the DIO group was significantly decreased, and the expression level of cleaved caspase-3 was significantly increased (p < 0.05). Compared with the control group, the Bcl-2 protein expression level in the testes of the DIO-R group significantly decreased (p < 0.05). However, the Bax expression level increased. Thus, the Bcl-2/Bax ratio significantly decreased (p < 0.01); however, the factor-related apoptosis (Fas), Fas ligand (FasLG), cleaved caspase-8, caspase-8, cleaved caspase-3, and caspase-3 protein expression levels significantly increased (p < 0.05). Compared with the DIO group, in DIO-R mice, the activities of ACE, ATF1, Bcl-2, and Bcl-2/Bax's spermatogenesis protein expression decreased, while the apoptosis-promoting protein expression significantly increased (p < 0.05). During the in vitro experiment, the late and early apoptotic ratio in the miR-34c over-expression group increased. MiR-34c over-expression enhanced the expression of apoptosis-related proteins Fas/FasLG and Bax/Bcl-2 while inhibiting the expression of ATF1 and the sperm-associated protein in GC-1 spg cells. DIO and DIO-R could harm sperm production. DIO-R could impair sperm production by inducing the miR-34c-activated apoptosis and spermatogenesis pathway, which may be different from that of DIO.


Sujet(s)
Apoptose , Alimentation riche en graisse , Souris de lignée C57BL , microARN , Obésité , Spermatogenèse , Spermatozoïdes , Animaux , Mâle , microARN/génétique , microARN/métabolisme , Spermatogenèse/génétique , Souris , Obésité/métabolisme , Obésité/génétique , Spermatozoïdes/métabolisme , Alimentation riche en graisse/effets indésirables , Lignée cellulaire
17.
Nat Commun ; 15(1): 5801, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987242

RÉSUMÉ

Obesity is a heritable disease, characterised by excess adiposity that is measured by body mass index (BMI). While over 1,000 genetic loci are associated with BMI, less is known about the genetic contribution to adiposity trajectories over adulthood. We derive adiposity-change phenotypes from 24.5 million primary-care health records in over 740,000 individuals in the UK Biobank, Million Veteran Program USA, and Estonian Biobank, to discover and validate the genetic architecture of adiposity trajectories. Using multiple BMI measurements over time increases power to identify genetic factors affecting baseline BMI by 14%. In the largest reported genome-wide study of adiposity-change in adulthood, we identify novel associations with BMI-change at six independent loci, including rs429358 (APOE missense variant). The SNP-based heritability of BMI-change (1.98%) is 9-fold lower than that of BMI. The modest genetic correlation between BMI-change and BMI (45.2%) indicates that genetic studies of longitudinal trajectories could uncover novel biology of quantitative traits in adulthood.


Sujet(s)
Adiposité , Indice de masse corporelle , Dossiers médicaux électroniques , Étude d'association pangénomique , Obésité , Polymorphisme de nucléotide simple , Humains , Adiposité/génétique , Mâle , Femelle , Obésité/génétique , Adulte d'âge moyen , Adulte , Sujet âgé , Royaume-Uni , Phénotype , Estonie , États-Unis , Prédisposition génétique à une maladie
18.
Cardiovasc Diabetol ; 23(1): 231, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965592

RÉSUMÉ

BACKGROUND: Associations between metabolic status and metabolic changes with the risk of cardiovascular outcomes have been reported. However, the role of genetic susceptibility underlying these associations remains unexplored. We aimed to examine how metabolic status, metabolic transitions, and genetic susceptibility collectively impact cardiovascular outcomes and all-cause mortality across diverse body mass index (BMI) categories. METHODS: In our analysis of the UK Biobank, we included a total of 481,576 participants (mean age: 56.55; male: 45.9%) at baseline. Metabolically healthy (MH) status was defined by the presence of < 3 abnormal components (waist circumstance, blood pressure, blood glucose, triglycerides, and high-density lipoprotein cholesterol). Normal weight, overweight, and obesity were defined as 18.5 ≤ BMI < 25 kg/m2, 25 ≤ BMI < 30 kg/m2, and BMI ≥ 30 kg/m2, respectively. Genetic predisposition was estimated using the polygenic risk score (PRS). Cox regressions were performed to evaluate the associations of metabolic status, metabolic transitions, and PRS with cardiovascular outcomes and all-cause mortality across BMI categories. RESULTS: During a median follow-up of 14.38 years, 31,883 (7.3%) all-cause deaths, 8133 (1.8%) cardiovascular disease (CVD) deaths, and 67,260 (14.8%) CVD cases were documented. Among those with a high PRS, individuals classified as metabolically healthy overweight had the lowest risk of all-cause mortality (hazard ratios [HR] 0.70; 95% confidence interval [CI] 0.65, 0.76) and CVD mortality (HR 0.57; 95% CI 0.50, 0.64) compared to those who were metabolically unhealthy obesity, with the beneficial associations appearing to be greater in the moderate and low PRS groups. Individuals who were metabolically healthy normal weight had the lowest risk of CVD morbidity (HR 0.54; 95% CI 0.51, 0.57). Furthermore, the inverse associations of metabolic status and PRS with cardiovascular outcomes and all-cause mortality across BMI categories were more pronounced among individuals younger than 65 years (Pinteraction < 0.05). Additionally, the combined protective effects of metabolic transitions and PRS on these outcomes among BMI categories were observed. CONCLUSIONS: MH status and a low PRS are associated with a lower risk of adverse cardiovascular outcomes and all-cause mortality across all BMI categories. This protective effect is particularly pronounced in individuals younger than 65 years. Further research is required to confirm these findings in diverse populations and to investigate the underlying mechanisms involved.


Sujet(s)
Indice de masse corporelle , Maladies cardiovasculaires , Cause de décès , Prédisposition génétique à une maladie , Hérédité multifactorielle , Obésité , Humains , Mâle , Adulte d'âge moyen , Femelle , Maladies cardiovasculaires/mortalité , Maladies cardiovasculaires/génétique , Maladies cardiovasculaires/diagnostic , Maladies cardiovasculaires/épidémiologie , Appréciation des risques , Études prospectives , Sujet âgé , Obésité/génétique , Obésité/diagnostic , Obésité/mortalité , Obésité/épidémiologie , Royaume-Uni/épidémiologie , Phénotype , Facteurs temps , Pronostic , Adulte , Obésité métaboliquement bénigne/diagnostic , Obésité métaboliquement bénigne/mortalité , Obésité métaboliquement bénigne/génétique , Obésité métaboliquement bénigne/épidémiologie , Facteurs de risque cardiométabolique , Facteurs de risque ,
19.
Nat Commun ; 15(1): 5919, 2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39004626

RÉSUMÉ

Pregnane X receptor (PXR) has been reported to regulate glycolipid metabolism. The dysfunction of intestinal barrier contributes to metabolic disorders. However, the role of intestinal PXR in metabolic diseases remains largely unknown. Here, we show that activation of PXR by tributyl citrate (TBC), an intestinal-selective PXR agonist, improves high fat diet (HFD)-induced obesity. The metabolic benefit of intestinal PXR activation is associated with upregulation of ß-1,3 galactosyltransferase 5 (B3galt5). Our results reveal that B3galt5 mainly expresses in the intestine and is a direct PXR transcriptional target. B3galt5 knockout exacerbates HFD-induced obesity, insulin resistance and inflammation. Mechanistically, B3galt5 is essential to maintain the integrity of intestinal mucus barrier. B3galt5 ablation impairs the O-glycosylation of mucin2, destabilizes the mucus layer, and increases intestinal permeability. Furthermore, B3galt5 deficiency abolishes the beneficial effect of intestinal PXR activation on metabolic disorders. Our results suggest the intestinal-selective PXR activation regulates B3galt5 expression and maintains metabolic homeostasis, making it a potential therapeutic strategy in obesity.


Sujet(s)
Alimentation riche en graisse , Galactosyltransferases , Insulinorésistance , Muqueuse intestinale , Souris de lignée C57BL , Souris knockout , Obésité , Récepteur du prégnane X , Animaux , Obésité/métabolisme , Obésité/génétique , Récepteur du prégnane X/métabolisme , Récepteur du prégnane X/génétique , Galactosyltransferases/métabolisme , Galactosyltransferases/génétique , Souris , Alimentation riche en graisse/effets indésirables , Muqueuse intestinale/métabolisme , Mâle , Intestins , Humains
20.
J Clin Invest ; 134(14)2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39007272

RÉSUMÉ

A growing body of research has identified circadian-rhythm disruption as a risk factor for metabolic health. However, the underlying biological basis remains complex, and complete molecular mechanisms are unknown. There is emerging evidence from animal and human research to suggest that the expression of core circadian genes, such as circadian locomotor output cycles kaput gene (CLOCK), brain and muscle ARNT-Like 1 gene (BMAL1), period (PER), and cyptochrome (CRY), and the consequent expression of hundreds of circadian output genes are integral to the regulation of cellular metabolism. These circadian mechanisms represent potential pathophysiological pathways linking circadian disruption to adverse metabolic health outcomes, including obesity, metabolic syndrome, and type 2 diabetes. Here, we aim to summarize select evidence from in vivo animal models and compare these results with epidemiologic research findings to advance understanding of existing foundational evidence and potential mechanistic links between circadian disruption and altered clock gene expression contributions to metabolic health-related pathologies. Findings have important implications for the treatment, prevention, and control of metabolic pathologies underlying leading causes of death and disability, including diabetes, cardiovascular disease, and cancer.


Sujet(s)
Protéines CLOCK , Rythme circadien , Diabète de type 2 , Humains , Animaux , Rythme circadien/génétique , Protéines CLOCK/génétique , Protéines CLOCK/métabolisme , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Obésité/génétique , Obésité/métabolisme , Syndrome métabolique X/génétique , Syndrome métabolique X/métabolisme , Horloges circadiennes/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...